50 results on '"Duijf, Pascal HG"'
Search Results
2. NDRG1 is a prognostic biomarker in breast cancer and breast cancer brain metastasis
- Author
-
Joshi, Vaibhavi, primary, Stacey, Andrew, additional, Feng, Yufan, additional, Kalita‐de Croft, Priyakshi, additional, Duijf, Pascal HG, additional, Simpson, Peter T, additional, Lakhani, Sunil R, additional, and McCart Reed, Amy E, additional
- Published
- 2024
- Full Text
- View/download PDF
3. Stochastic epithelial-mesenchymal transitions diversify non-cancerous lung cell behaviours
- Author
-
Bhatia, Sugandha, primary, Gunter, Jennifer H, additional, Burgess, Joshua T, additional, Adams, Mark N, additional, O'Byrne, Kenneth, additional, Thompson, Erik W, additional, and Duijf, Pascal HG, additional
- Published
- 2023
- Full Text
- View/download PDF
4. CEP55 is a determinant of cell fate during perturbed mitosis in breast cancer
- Author
-
Kalimutho, Murugan, Sinha, Debottam, Jeffery, Jessie, Nones, Katia, Srihari, Sriganesh, Fernando, Winnie C, Duijf, Pascal HG, Vennin, Claire, Raninga, Prahlad, Nanayakkara, Devathri, Mittal, Deepak, Saunus, Jodi M, Lakhani, Sunil R, López, J Alejandro, Spring, Kevin J, Timpson, Paul, Gabrielli, Brian, Waddell, Nicola, and Khanna, Kum Kum
- Published
- 2018
- Full Text
- View/download PDF
5. Corrigendum to “MicroRNAs in cancer cell death pathways: Apoptosis and necroptosis” [Free Radic. Biol. Med. 139 (2019) 1–15]
- Author
-
Shirjang, Solmaz, primary, Mansoori, Behzad, additional, Asghari, Samira, additional, Duijf, Pascal HG., additional, Mohammadi, Ali, additional, Gjerstorff, Morten, additional, and Baradaran, Behzad, additional
- Published
- 2020
- Full Text
- View/download PDF
6. Multi-Omics Characterization of the Spontaneous Mesenchymal–Epithelial Transition in the PMC42 Breast Cancer Cell Lines
- Author
-
Bhatia, Sugandha, primary, Monkman, James, additional, Blick, Tony, additional, Duijf, Pascal HG, additional, Nagaraj, Shivashankar H., additional, and Thompson, Erik W., additional
- Published
- 2019
- Full Text
- View/download PDF
7. Cover Image, Volume 233, Number 3, March 2018
- Author
-
Ghasabi, Mehri, primary, Mansoori, Behzad, additional, Mohammadi, Ali, additional, Duijf, Pascal HG, additional, Shomali, Navid, additional, Shirafkan, Naghmeh, additional, Mokhtarzadeh, Ahad, additional, and Baradaran, Behzad, additional
- Published
- 2018
- Full Text
- View/download PDF
8. MicroRNAs in cancer drug resistance: Basic evidence and clinical applications
- Author
-
Ghasabi, Mehri, primary, Mansoori, Behzad, additional, Mohammadi, Ali, additional, Duijf, Pascal HG, additional, Shomali, Navid, additional, Shirafkan, Naghmeh, additional, Mokhtarzadeh, Ahad, additional, and Baradaran, Behzad, additional
- Published
- 2018
- Full Text
- View/download PDF
9. CEP 55 is a determinant of cell fate during perturbed mitosis in breast cancer
- Author
-
Kalimutho, Murugan, primary, Sinha, Debottam, additional, Jeffery, Jessie, additional, Nones, Katia, additional, Srihari, Sriganesh, additional, Fernando, Winnie C, additional, Duijf, Pascal HG, additional, Vennin, Claire, additional, Raninga, Prahlad, additional, Nanayakkara, Devathri, additional, Mittal, Deepak, additional, Saunus, Jodi M, additional, Lakhani, Sunil R, additional, López, J Alejandro, additional, Spring, Kevin J, additional, Timpson, Paul, additional, Gabrielli, Brian, additional, Waddell, Nicola, additional, and Khanna, Kum Kum, additional
- Published
- 2018
- Full Text
- View/download PDF
10. MicroRNAs in cancer drug resistance: Basic evidence and clinical applications.
- Author
-
Ghasabi, Mehri, Mansoori, Behzad, Mohammadi, Ali, Duijf, Pascal HG, Shomali, Navid, Shirafkan, Naghmeh, Mokhtarzadeh, Ahad, and Baradaran, Behzad
- Subjects
MICRORNA ,DRUG resistance in cancer cells ,CANCER treatment ,CANCER chemotherapy ,MONOCLONAL antibodies - Abstract
Development of drug resistance has considerably limited the efficacy of cancer treatments, including chemotherapy and targeted therapies. Hence, understanding the molecular mechanisms underpinning the innate or the acquired resistance to these therapies is critical to improve drug efficiency and clinical outcomes. Several studies have implicated microRNAs (miRNA) in this process. MiRNAs repress gene expression by specific binding to complementary sequences in the 3' region of target messenger RNAs (mRNAs), followed by target mRNA degradation or blocked translation. By targeting molecules specific to a particular pathway within tumor cells, the new generation of cancer treatment strategies has shown significant advantages over conventional chemotherapy. However, the long‐term efficacy of targeted therapies often remains poor, because tumor cells develop resistance to such therapeutics. Targeted therapies often involve monoclonal antibodies (mAbs), such as those blocking the ErB/HER tyrosine kinases, epidermal growth factor receptor (cetuximab) and HER2 (trastuzumab), and those inhibiting vascular endothelial growth factor receptor signaling (e.g., bevacizumab). Even though these are among the most used agents in tumor medicine, clinical response to these drugs is reduced due to the emergence of drug resistance as a result of toxic effects in the tumor microenvironment. Research on different types of human cancers has revealed that aberrant expression of miRNAs promotes resistance to the aforementioned drugs. In this study, we review the mechanisms of tumor cell resistance to mAb therapies and the role of miRNAs therein. Emerging treatment strategies combine therapies using innovative miRNA mimics or antagonizers with conventional approaches to maximize outcomes of patients with cancer. This literature review discuss the role of microRNAs in cancer cell resistance to targeted therapies. In addition, it discuss the molecular mechanisms underlying the development of therapeutic drug resistance based on basic evidence and clinical applications. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
11. Abstract 3586: Overexpression of Emi1 causes chromosomal instability and cancer
- Author
-
Vaidyanathan, Srividya, primary, Cato, Kathleen, additional, Pavey, Sandra, additional, Haass, Nikolas K., additional, Gabrielli, Brian G., additional, and Duijf., Pascal HG, additional
- Published
- 2016
- Full Text
- View/download PDF
12. Cover Image, Volume 233, Number 3, March 2018.
- Author
-
Ghasabi, Mehri, Mansoori, Behzad, Mohammadi, Ali, Duijf, Pascal HG, Shomali, Navid, Shirafkan, Naghmeh, Mokhtarzadeh, Ahad, and Baradaran, Behzad
- Subjects
MICRORNA ,DRUG resistance in cancer cells - Abstract
Cover: The cover image is based on the Review Article MicroRNAs in cancer drug resistance: Basic evidence and clinical applications by Mehri Ghasabi et al., DOI: 10.1002/jcp.26810. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
13. Alpha-B-Crystallin overexpression is sufficient to promote tumorigenesis and metastasis in mice
- Author
-
Kum Kum Khanna, Behnam Rashidieh, Amanda Bain, Simon Tria, Sowmya Sharma, Cameron Stewart, Jacinta Simmons, Pirjo Apaja, Pascal Duijf, John Finnie, Rashidieh, Behnam, Bain, Amanda Louise, Tria, Simon Manuel, Sharma, Sowmya, Stewart, Cameron Allan, Simmons, Jacinta Ley, Apaja, Pirjo M, Duijf, Pascal HG, Finnie, John, and Khanna, Kum Kum
- Subjects
Cancer Research ,tumorigenesis ,angiogenesis ,Oncology ,metastasis ,Hematology ,heat-shock protein ,Cryab mouse model - Abstract
Background αB-Crystallin is a heat shock chaperone protein which binds to misfolded proteins to prevent their aggregation. It is overexpressed in a wide-variety of cancers. Previous studies using human cancer cell lines and human xenograft models have suggested potential tumor promoter (oncogene) roles for αB-Crystallin in a wide-spectrum of cancers. Methods To determine the causal relationship between CRYAB overexpression and cancer, we generated a Cryab overexpression knock-in mouse model and monitor them for development of spontaneous and carcinogen (DMBA)-induced tumorigenesis. In order to investigate the mechanism of malignancies observed in this model multiple techniques were used such as immunohistochemical characterizations of tumors, bioinformatics analysis of publically available human tumor datasets, and generation of mouse embryonic fibroblasts (MEFs) for in vitro assays (clonogenic survival and migration assays and proteome analysis by mass-spectrometry). Results This model revealed that constitutive overexpression of Cryab results in the formation of a variety of lethal spontaneous primary and metastatic tumors in mice. In vivo, the overexpression of Cryab correlated with the upregulation of epithelial-to-mesenchymal (EMT) markers, angiogenesis and some oncogenic proteins including Basigin. In vitro, using E1A/Ras transformed MEFs, we observed that the overexpression of Cryab led to the promotion of cell survival via upregulation of Akt signaling and downregulation of pro-apoptotic pathway mediator JNK, with subsequent attenuation of apoptosis as assessed by cleaved caspase-3 and Annexin V staining. Conclusions Overall, through the generation and characterization of Cryab overexpression model, we provide evidence supporting the role of αB-Crystallin as an oncogene, where its upregulation is sufficient to induce tumors, promote cell survival and inhibit apoptosis.
- Published
- 2023
14. hSSB1 (NABP2/OBFC2B) modulates the DNA damage and androgen-induced transcriptional response in prostate cancer
- Author
-
Mark N. Adams, Laura V. Croft, Aaron Urquhart, Mohamed Ashick Mohamed Saleem, Anja Rockstroh, Pascal H. G. Duijf, Patrick B. Thomas, Genevieve P. Ferguson, Idris Mohd Najib, Esha T. Shah, Emma Bolderson, Shivashankar Nagaraj, Elizabeth D. Williams, Colleen C. Nelson, Kenneth J. O'Byrne, Derek J. Richard, Adams, Mark N, Croft, Laura V, Urquhart, Aaron, Saleem, Mohamed Ashick Mohamed, Rockstroh, Anja, Duijf, Pascal HG, Thomas, Patrick B, Ferguson, Genevieve P, Najib, Idris Mohd, Shah, Esha T, Bolderson, Emma, Nagaraj, Shivashankar, Williams, Elizabeth D, Nelson, Colleen C, O'Byrne, Kenneth J, and Richard, Derek J
- Subjects
radiation ,Oncology ,Urology ,androgen receptor ,DNA damage ,hSSB1/NABP2 ,prostate cancer - Abstract
Background: Activation and regulation of androgen receptor (AR) signaling and the DNA damage response impact the prostate cancer (PCa) treatment modalities of androgen deprivation therapy (ADT) and radiotherapy. Here, we have evaluated a role for human single-strand binding protein 1 (hSSB1/NABP2) in modulation of the cellular response to androgens and ionizing radiation (IR). hSSB1 has defined roles in transcription and maintenance of genome stability, yet little is known about this protein in PCa. Methods: We correlated hSSB1 with measures of genomic instability across available PCa cases from The Cancer Genome Atlas (TCGA). Microarray and subsequent pathway and transcription factor enrichment analysis were performed on LNCaP and DU145 prostate cancer cells. Results: Our data demonstrate that hSSB1 expression in PCa correlates with measures of genomic instability including multigene signatures and genomic scars that are reflective of defects in the repair of DNA double-strand breaks via homologous recombination. In response to IR-induced DNA damage, we demonstrate that hSSB1 regulates cellular pathways that control cell cycle progression and the associated checkpoints. In keeping with a role for hSSB1 in transcription, our analysis revealed that hSSB1 negatively modulates p53 and RNA polymerase II transcription in PCa. Of relevance to PCa pathology, our findings highlight a transcriptional role for hSSB1 in regulating the androgen response. We identified that AR function is predicted to be impacted by hSSB1 depletion, whereby this protein is required to modulate AR gene activity in PCa. Conclusions: Our findings point to a key role for hSSB1 in mediating the cellular response to androgen and DNA damage via modulation of transcription. Exploiting hSSB1 in PCa might yield benefits as a strategy to ensure a durable response to ADT and/or radiotherapy and improved patient outcomes. Refereed/Peer-reviewed
- Published
- 2023
15. Noncoding RNAs in oral cancer
- Author
-
Jaikrishna Balakittnen, Chameera Ekanayake Weeramange, Daniel F. Wallace, Pascal H. G. Duijf, Alexandre S. Cristino, Liz Kenny, Sarju Vasani, Chamindie Punyadeera, Balakittnen, Jaikrishna, Weeramange, Chameera Ekanayake, Wallace, Daniel F, Duijf, Pascal HG, Cristino, Alexandre S, Kenny, Liz, Vasani, Sarju, and Punyadeera, Chamindie
- Subjects
diagnosis ,therapeutics ,prognosis ,oral cancer ,Molecular Biology ,Biochemistry ,noncoding RNA - Abstract
Oral cancer (OC) is the most prevalent subtype of cancer arising in the head and neck region. OC risk is mainly attributed to behavioral risk factors such as exposure to tobacco and excessive alcohol consumption, and a lesser extent to viral infections such as human papillomaviruses and Epstein-Barr viruses. In addition to these acquired risk factors, heritable genetic factors have shown to be associated with OC risk. Despite the high incidence, biomarkers for OC diagnosis are lacking and consequently, patients are often diagnosed in advanced stages. This delay in diagnosis is reflected by poor overall outcomes of OC patients, where 5-year overall survival is around 50%. Among the biomarkers proposed for cancer detection, noncoding RNA (ncRNA) can be considered as one of the most promising categories of biomarkers due to their role in virtually all cellular processes. Similar to other cancer types, changes in expressions of ncRNAs have been reported in OC and a number of ncRNAs have diagnostic, prognostic, and therapeutic potential. Moreover, some ncRNAs are capable of regulating gene expression by various mechanisms. Therefore, elucidating the current literature on the four main types of ncRNAs namely, microRNA, lncRNA, snoRNA, piwi-RNA, and circular RNA in the context of OC pathogenesis is timely and would enable further improvements and innovations in diagnosis, prognosis, and treatment of OC. This article is categorized under: RNA in Disease and DevelopmentRNA in Disease RNA in Disease and DevelopmentRNA in Development.
- Published
- 2022
16. Cep55 overexpression promotes genomic instability and tumorigenesis in mice
- Author
-
Purba Nag, Meaghan Wall, Andrew Burgess, Debottam Sinha, John W. Finnie, Kum Kum Khanna, Murugan Kalimutho, Devathri Nanayakkara, Veronique A. J. Smits, Goutham Subramanian, Prahlad V. Raninga, Pascal H.G. Duijf, Amanda L. Bain, Sinha, Debottam, Nag, Purba, Nanayakkara, Devathri, Duijf, Pascal HG, Burgess, Andrew, Raninga, Prahlad, Smits, Veronique AJ, Bain, Amanda L, Subramanian, Goutham, Wall, Meaghan, Finnie, John W, Kalimutho, Murugan, and Khanna, Kum Kum
- Subjects
0301 basic medicine ,Genome instability ,Biopsy ,Gene Expression ,Medicine (miscellaneous) ,Cell Cycle Proteins ,medicine.disease_cause ,Microtubules ,Mice ,0302 clinical medicine ,Chromosome instability ,lcsh:QH301-705.5 ,Protein Stability ,Immunohistochemistry ,Phenotype ,Cell biology ,Mechanisms of disease ,Cell Transformation, Neoplastic ,030220 oncology & carcinogenesis ,Disease Susceptibility ,General Agricultural and Biological Sciences ,Signal Transduction ,Genetically modified mouse ,Genotype ,DNA damage ,Karyotype ,Mitosis ,Mice, Transgenic ,Biology ,Article ,Genomic Instability ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Stress, Physiological ,Biomarkers, Tumor ,medicine ,cancer ,Animals ,CEP55 expression ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Oncogenes ,Fibroblasts ,030104 developmental biology ,lcsh:Biology (General) ,Checkpoint Kinase 1 ,cells ,Lymph Nodes ,mutation ,Tumor Suppressor Protein p53 ,protein ,Carcinogenesis ,Proto-Oncogene Proteins c-akt - Abstract
High expression of centrosomal protein CEP55 has been correlated with clinico-pathological parameters across multiple human cancers. Despite significant in vitro studies and association of aberrantly overexpressed CEP55 with worse prognosis, its causal role in vivo tumorigenesis remains elusive. Here, using a ubiquitously overexpressing transgenic mouse model, we show that Cep55 overexpression causes spontaneous tumorigenesis and accelerates Trp53+/− induced tumours in vivo. At the cellular level, using mouse embryonic fibroblasts (MEFs), we demonstrate that Cep55 overexpression induces proliferation advantage by modulating multiple cellular signalling networks including the hyperactivation of the Pi3k/Akt pathway. Notably, Cep55 overexpressing MEFs have a compromised Chk1-dependent S-phase checkpoint, causing increased replication speed and DNA damage, resulting in a prolonged aberrant mitotic division. Importantly, this phenotype was rescued by pharmacological inhibition of Pi3k/Akt or expression of mutant Chk1 (S280A) protein, which is insensitive to regulation by active Akt, in Cep55 overexpressing MEFs. Moreover, we report that Cep55 overexpression causes stabilized microtubules. Collectively, our data demonstrates causative effects of deregulated Cep55 on genome stability and tumorigenesis which have potential implications for tumour initiation and therapy development., Sinha et al. demonstrate that overexpression of centrosomal protein Cep55 in mice is sufficient to cause a wide-spectrum of cancer via multiple mechanisms including hyperactivation of the Pi3k/Akt pathway, stabilized microtubules and a defective replication checkpoint response. These findings are relevant to human cancers as high CEP55 expression is associated with worse prognosis across multiple cancer types.
- Published
- 2020
- Full Text
- View/download PDF
17. miR-330 Regulates Colorectal Cancer Oncogenesis by Targeting BACH1
- Author
-
Behzad Baradaran, Neda Shajari, Behzad Mansoori, Ali Mohammadi, Katayoon Nofouzi, Pascal H.G. Duijf, Fereydoon Abedi Gaballu, Solmaz Shirjang, Souzan Najafi, Shirjang, Solmaz, Mansoori, Behzad, Mohammadi, Ali, Shajari, Neda, Duijf, Pascal HG, Najafi, Souzan, Gaballu, Fereydoon Abedi, Nofouzi, Katayoon, and Baradaran, Behzad
- Subjects
Colorectal cancer ,medicine.medical_treatment ,proliferation ,Pharmaceutical Science ,colorectal cancer ,BACH1 ,medicine.disease_cause ,CXCR4 ,miR-330 ,Targeted therapy ,VEGFR ,03 medical and health sciences ,mir-330 ,0302 clinical medicine ,microRNA ,medicine ,Gene silencing ,General Pharmacology, Toxicology and Pharmaceutics ,030304 developmental biology ,0303 health sciences ,MMP9 ,business.industry ,bach1 ,lcsh:RM1-950 ,vegfr ,Oncomir ,medicine.disease ,digestive system diseases ,Blot ,lcsh:Therapeutics. Pharmacology ,030220 oncology & carcinogenesis ,Cancer research ,mmp9 ,Carcinogenesis ,business ,cxcr4 ,Research Article - Abstract
Purpose: Based on WHO report, colorectal cancer (CRC) is the second cause of death among patients with cancer worldwide. Dysregulation of miRNAs expressions has been demonstrated in different human cancers, especially CRC. Studies have shown that miR-330 could act as both TS-miR and/or oncomiR in different types of cancers. BACH1 is also identified as a transcription factor, which is involved in ontogenesis. In this study, we evaluated the CRC suppression via silencing of BACH1 by small silencer molecule called miR-330.Methods:Firstly, we analyzed the BACH1, miR-330-3p and miR-330-5p expressions according to the colon adenocarcinoma (COAD) and rectal adenocarcinoma (READ) project established from a patient of the colon and rectal cancer patients in The Cancer Genome Atlas (TCGA) database. The targeting of BACH1 via miR-330 in human CRC cells was evaluated by Vejnar bioinformatics methods, and confirmed by qRT-PCR and western blot analysis. Proliferation was performed by MTT assay. The MMP9, CXCR4, and VEGFR proteins were measured by western blotting.Results:The analysis of BACH1, miR-330-3p, and miR-330-5p expressions according to the COAD and READ projects showed that BACH1 was overexpressed, but miR-330-3p and miR330-5p were reduced in CRC tumors compared to normal controls. The miR-330 induction prevented proliferation of CRC cell by targeting BACH1 mRNA, which represses MMP9, C-X-C chemokine receptor type 4 (CXCR4), and vascular endothelial growth factor receptor (VEGFR) proteins expressions.Conclusion:Our results suggested that BACH1 is a potential target for miR-330 in CRC cells. The miR-330 induction inhibits CRC cells proliferation by suppressing BACH1 expression in posttranscriptional level. It was suggested that targeting of BACH1 via miRNA such as miR-330 could be a valid strategy in the field of CRC targeted therapy via modulating the oncogenic signaling pathway.
- Published
- 2020
- Full Text
- View/download PDF
18. Chromosome arm aneuploidies shape tumour evolution and drug response
- Author
-
Harald Oey, J. Lynn Fink, Ankit Shukla, Kum Kum Khanna, Eloise Dray, Jonathan Ellis, Alexandre S. Cristino, Thu H.M. Nguyen, John P. Grady, Dirk P. Kroese, Lutz Krause, Pascal H.G. Duijf, Sarat B. Moka, Shukla, Ankit, Nguyen, Thu HM, Moka, Sarat B, Ellis, Jonathan J, Grady, John P, Oey, Harald, Cristino, Alexandre S, Khanna, Kum Kum, Kroese, Dirk P, Krause, Lutz, Dray, Eloise, Fink, J Lynn, and Duijf, Pascal HG
- Subjects
0301 basic medicine ,Mutation rate ,General Physics and Astronomy ,Aneuploidy ,Kaplan-Meier Estimate ,Drug resistance ,Metastasis ,Machine Learning ,0302 clinical medicine ,Mutation Rate ,Neoplasms ,Chromosomes, Human ,Medicine ,chromosome ,lcsh:Science ,Multidisciplinary ,drug ,Prognosis ,Cancer therapeutic resistance ,machine learning ,030220 oncology & carcinogenesis ,Chromosome Arm ,Systems biology ,tumor ,Science ,Models, Biological ,Article ,General Biochemistry, Genetics and Molecular Biology ,developmental biology ,03 medical and health sciences ,Cell Line, Tumor ,evolution ,mental disorders ,Humans ,Chemotherapy ,cancer ,cardiovascular diseases ,Stochastic Processes ,business.industry ,Chromosome ,Cancer ,nutritional and metabolic diseases ,General Chemistry ,medicine.disease ,030104 developmental biology ,Drug Resistance, Neoplasm ,Pharmacogenomics ,Cancer research ,lcsh:Q ,cell component ,business - Abstract
Chromosome arm aneuploidies (CAAs) are pervasive in cancers. However, how they affect cancer development, prognosis and treatment remains largely unknown. Here, we analyse CAA profiles of 23,427 tumours, identifying aspects of tumour evolution including probable orders in which CAAs occur and CAAs predicting tissue-specific metastasis. Both haematological and solid cancers initially gain chromosome arms, while only solid cancers subsequently preferentially lose multiple arms. 72 CAAs and 88 synergistically co-occurring CAA pairs multivariately predict good or poor survival for 58% of 6977 patients, with negligible impact of whole-genome doubling. Additionally, machine learning identifies 31 CAAs that robustly alter response to 56 chemotherapeutic drugs across cell lines representing 17 cancer types. We also uncover 1024 potential synthetic lethal pharmacogenomic interactions. Notably, in predicting drug response, CAAs substantially outperform mutations and focal deletions/amplifications combined. Thus, CAAs predict cancer prognosis, shape tumour evolution, metastasis and drug response, and may advance precision oncology., Chromosome arm-level aneuploidies (CAAs) are frequently observed in cancer. Here, the authors analyse CAA landscapes across different tumour types, relating these chromosome arm gains and losses to tumour evolution, metastasis, patient survival and response to a range of anti-cancer therapies.
- Published
- 2020
19. Identification of common and distinct pathways in inflammatory bowel disease and colorectal cancer: a hypothesis based on weighted gene co-expression network analysis
- Author
-
Afshin Derakhshani, Darya Javadrashid, Nima Hemmat, Antoine Dufour, Antonio Giovanni Solimando, Mahdi Abdoli Shadbad, Pascal H. G. Duijf, Oronzo Brunetti, Nicola Silvestris, Behzad Baradaran, Derakhshani, Afshin, Javadrashid, Darya, Hemmat, Nima, Dufour, Antoine, Solimando, Antonio Giovanni, Abdoli Shadbad, Mahdi, Duijf, Pascal HG, Brunetti, Oronzo, Silvestris, Nicola, and Baradaran, Behzad
- Subjects
inflammatory bowel disease ,WGCNA ,pathogenesis ,Genetics ,Molecular Medicine ,colorectal cancer ,systems biology ,digestive system diseases ,Genetics (clinical) - Abstract
Patients with inflammatory bowel disease (IBD), including ulcerative colitis and Crohn’s disease, are at higher risk to develop colorectal cancer (CRC). However, the underlying mechanisms of this predisposition remain elusive. We performed in-depth comparative computational analyses to gain new insights, including weighted gene co-expression network analysis (WGCNA) and gene ontology and pathway enrichment analyses, using gene expression datasets from IBD and CRC patients. When individually comparing IBD and CRC to normal control samples, we identified clusters of highly correlated genes, differentially expressed genes, and module-trait associations specific for each disease. When comparing IBD to CRC, we identified common hub genes and commonly enriched pathways. Most notably, IBD and CRC share significantly increased expression of five genes (MMP10, LCN2, REG1A, REG3A, and DUOX2), enriched inflammatory and neutrophil activation pathways and, most notably, highly significant enrichment of IL-4 and IL-13 signaling. Thus, our work expands our knowledge about the intricate relationship between IBD and CRC development and provides new rationales for developing novel therapeutic strategies.
- Published
- 2022
20. Targeting BRF2 in Cancer Using Repurposed Drugs
- Author
-
Behnam Rashidieh, Simon Manuel Tria, Maryam Molakarimi, Rachael C. Adams, Mathew J. K. Jones, Hein Truong, Murugan Kalimutho, Pascal H.G. Duijf, Sriganesh Srihari, Kum Kum Khanna, Ammar Mohseni, Rashidieh, Behnam, Molakarimi, Maryam, Mohseni, Ammar, Tria, Simon Manuel, Truong, Hein, Srihari, Sriganesh, Adams, Rachael C, Jones, Mathew, Duijf, Pascal HG, Kalimutho, Murugan, and Khanna, Kum Kum
- Subjects
0301 basic medicine ,Cancer Research ,DNA damage ,medicine.disease_cause ,Article ,03 medical and health sciences ,0302 clinical medicine ,medicine ,cancer ,Transcription factor ,RC254-282 ,Bexarotene ,drug repurposing ,Chemistry ,bexarotene ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cancer ,medicine.disease ,Drug repositioning ,030104 developmental biology ,molecular dynamics simulation ,Oncology ,Apoptosis ,BRF2 ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Oxidative stress ,medicine.drug - Abstract
Simple Summary BRF2, a subunit of the RNA polymerase III transcription complex, is upregulated in a wide variety of cancers and is a potential therapeutic target; however, no effective drugs are available to target BRF2. The upregulation of BRF2 in cancer cells confers survival via the prevention of oxidative stress-induced apoptosis. In this manuscript, we report the identification of potential BRF2 inhibitors through in silico drug repurposing screening. We further characterized bexarotene as a hit compound for the development of selective BRF2 inhibitors and provide experimental validation to support the repurposing of this FDA-approved drug as an agent to reduce the cellular levels of ROS and consequent BRF2 expression in cancers with elevated levels of oxidative stress. Abstract The overexpression of BRF2, a selective subunit of RNA polymerase III, has been shown to be crucial in the development of several types of cancers, including breast cancer and lung squamous cell carcinoma. Predominantly, BRF2 acts as a central redox-sensing transcription factor (TF) and is involved in rescuing oxidative stress (OS)-induced apoptosis. Here, we showed a novel link between BRF2 and the DNA damage response. Due to the lack of BRF2-specific inhibitors, through virtual screening and molecular dynamics simulation, we identified potential drug candidates that interfere with BRF2-TATA-binding Protein (TBP)-DNA complex interactions based on binding energy, intermolecular, and torsional energy parameters. We experimentally tested bexarotene as a potential BRF2 inhibitor. We found that bexarotene (Bex) treatment resulted in a dramatic decline in oxidative stress and Tert-butylhydroquinone (tBHQ)-induced levels of BRF2 and consequently led to a decrease in the cellular proliferation of cancer cells which may in part be due to the drug pretreatment-induced reduction of ROS generated by the oxidizing agent. Our data thus provide the first experimental evidence that BRF2 is a novel player in the DNA damage response pathway and that bexarotene can be used as a potential inhibitor to treat cancers with the specific elevation of oxidative stress.
- Published
- 2021
- Full Text
- View/download PDF
21. Genome instability and pressure on non-homologous end joining drives chemotherapy resistance via a DNA repair crisis switch in triple negative breast cancer
- Author
-
Ambber Ward, Mark N. Adams, Romy Van Oosterhout, Idris Mohd Najib, Derek J. Richard, Pascal H.G. Duijf, Ekaterina Ivanova, Kenneth J. O'Byrne, Jason Sang Hun Lee, Scott W. Morrical, Martin C. Sadowski, Adrian P. Wiegmans, Greg Kelly, Wiegmans, Adrian P, Ward, Ambber, Ivanova, Ekaterina, Duijf, Pascal HG, Adams, Mark N, Najib, Idris Mohd, Van Oosterhout, Romy, Sadowski, Martin C, Kelly, Greg, Morrical, Scott W, O'Byrne, Ken, Lee, Jason S, and Richard, Derek J
- Subjects
0301 basic medicine ,Genome instability ,AcademicSubjects/SCI01140 ,AcademicSubjects/SCI01060 ,DNA damage ,DNA repair ,protein p53 ,AcademicSubjects/SCI00030 ,RAD51 ,610 Medicine & health ,Standard Article ,Biology ,DNA dependent protein kinase ,AcademicSubjects/SCI01180 ,doxorubicin ,03 medical and health sciences ,0302 clinical medicine ,docetaxel ,tumor protein p73 ,Triple-negative breast cancer ,DNA ,General Medicine ,Rad51 protein ,Homologous Recombination Pathway ,Non-homologous end joining ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,570 Life sciences ,biology ,AcademicSubjects/SCI00980 ,Homologous recombination - Abstract
Chemotherapy is used as a standard-of-care against cancers that display high levels of inherent genome instability. Chemotherapy induces DNA damage and intensifies pressure on the DNA repair pathways that can lead to deregulation. There is an urgent clinical need to be able to track the emergence of DNA repair driven chemotherapy resistance and tailor patient staging appropriately. There have been numerous studies into chemoresistance but to date no study has elucidated in detail the roles of the key DNA repair components in resistance associated with the frontline clinical combination of anthracyclines and taxanes together. In this study, we hypothesized that the emergence of chemotherapy resistance in triple negative breast cancer was driven by changes in functional signaling in the DNA repair pathways. We identified that consistent pressure on the non-homologous end joining pathway in the presence of genome instability causes failure of the key kinase DNA-PK, loss of p53 and compensation by p73. In-turn a switch to reliance on the homologous recombination pathway and RAD51 recombinase occurred to repair residual double strand DNA breaks. Further we demonstrate that RAD51 is an actionable target for resensitization to chemotherapy in resistant cells with a matched gene expression profile of resistance highlighted by homologous recombination in clinical samples.
- Published
- 2021
- Full Text
- View/download PDF
22. Dysregulated G2 phase checkpoint recovery pathway reduces DNA repair efficiency and increases chromosomal instability in a wide range of tumours
- Author
-
Martina Proctor, Mark N. Adams, Alexander J. Stevenson, Brian Gabrielli, Madushan Fernando, Anna Ehmann, Shivam Vora, Dubravka Skalamera, Pascal H.G. Duijf, Fernando, Madushan, Duijf, Pascal HG, Proctor, Martina, Stevenson, Alexander J, Ehmann, Anna, Vora, Shivam, Skalamera, Dubravka, Adams, Mark, and Gabrielli, Brian
- Subjects
0301 basic medicine ,Genome instability ,Cancer Research ,Cell cycle checkpoint ,ATM protein ,DNA repair ,DNA damage ,DNA replication ,Biology ,Gene mutation ,etoposide ,PLK1 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Chromosome instability ,Molecular Biology ,RC254-282 ,polo like kinase 1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Oncogenes ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Homologous recombination ,ATR protein - Abstract
Defective DNA repair is being demonstrated to be a useful target in cancer treatment. Currently, defective repair is identified by specific gene mutations, however defective repair is a common feature of cancers without these mutations. DNA damage triggers cell cycle checkpoints that are responsible for co-ordinating cell cycle arrest and DNA repair. Defects in checkpoint signalling components such as ataxia telangiectasia mutated (ATM) occur in a low proportion of cancers and are responsible for reduced DNA repair and increased genomic instability. Here we have investigated the AURKA-PLK1 cell cycle checkpoint recovery pathway that is responsible for exit from the G2 phase cell cycle checkpoint arrest. We demonstrate that dysregulation of PP6 and AURKA maintained elevated PLK1 activation to promote premature exit from only ATM, and not ATR-dependent checkpoint arrest. Surprisingly, depletion of the B55α subunit of PP2A that negatively regulates PLK1 was capable of overcoming ATM and ATR checkpoint arrests. Dysregulation of the checkpoint recovery pathway reduced S/G2 phase DNA repair efficiency and increased genomic instability. We found a strong correlation between dysregulation of the PP6-AURKA-PLK1-B55α checkpoint recovery pathway with signatures of defective homologous recombination and increased chromosomal instability in several cancer types. This work has identified an unrealised source of G2 phase DNA repair defects and chromosomal instability that are likely to be sensitive to treatments targeting defective repair.
- Published
- 2021
- Full Text
- View/download PDF
23. Elevating CDCA3 levels in non-small cell lung cancer enhances sensitivity to platinum-based chemotherapy
- Author
-
Kenneth J. O'Byrne, Pascal H.G. Duijf, Mark N. Adams, Sam Beard, Christopher Molloy, Eric D. Boittier, Derek J. Richard, Joshua T. Burgess, Emma Bolderson, Katherine B. Sahin, Neha S. Gandhi, Katrina Kildey, Esha T. Shah, Amila Suraweera, Kildey, Katrina, Gandhi, Neha S, Sahin, Katherine B, Shah, Esha T, Boittier, Eric, Duijf, Pascal HG, Molloy, Christopher, Burgess, Joshua T, Beard, Sam, Bolderson, Emma, Suraweera, Amila, Richard, Derek J, O'Byrne, Kenneth J, and Adams, Mark N
- Subjects
0301 basic medicine ,Lung Neoplasms ,medicine.medical_treatment ,Medicine (miscellaneous) ,Cell Cycle Proteins ,Biomarkers, Pharmacological ,chemistry.chemical_compound ,Prognostic markers ,0302 clinical medicine ,Maintenance therapy ,Carcinoma, Non-Small-Cell Lung ,Databases, Genetic ,CDCA3 protein ,Medicine ,platinum ,Biology (General) ,Casein Kinase II ,biology ,Cell Cycle ,Cadherins ,Ubiquitin ligase ,030220 oncology & carcinogenesis ,Casein kinase 2 ,General Agricultural and Biological Sciences ,medicine.drug ,QH301-705.5 ,General Biochemistry, Genetics and Molecular Biology ,Article ,Genomic Instability ,03 medical and health sciences ,Drug Therapy ,Antigens, CD ,Cell Line, Tumor ,Humans ,Lung cancer ,Cell Proliferation ,Platinum ,Cisplatin ,Chemotherapy ,non small cell lung cancer ,business.industry ,Immunotherapy ,medicine.disease ,Carboplatin ,030104 developmental biology ,chemistry ,Drug Resistance, Neoplasm ,biology.protein ,Cancer research ,business ,Non-small-cell lung cancer - Abstract
Platinum-based chemotherapy remains the cornerstone of treatment for most non-small cell lung cancer (NSCLC) cases either as maintenance therapy or in combination with immunotherapy. However, resistance remains a primary issue. Our findings point to the possibility of exploiting levels of cell division cycle associated protein-3 (CDCA3) to improve response of NSCLC tumours to therapy. We demonstrate that in patients and in vitro analyses, CDCA3 levels correlate with measures of genome instability and platinum sensitivity, whereby CDCA3high tumours are sensitive to cisplatin and carboplatin. In NSCLC, CDCA3 protein levels are regulated by the ubiquitin ligase APC/C and cofactor Cdh1. Here, we identified that the degradation of CDCA3 is modulated by activity of casein kinase 2 (CK2) which promotes an interaction between CDCA3 and Cdh1. Supporting this, pharmacological inhibition of CK2 with CX-4945 disrupts CDCA3 degradation, elevating CDCA3 levels and increasing sensitivity to platinum agents. We propose that combining CK2 inhibitors with platinum-based chemotherapy could enhance platinum efficacy in CDCA3low NSCLC tumours and benefit patients., Kildey et al find that high levels of mitotic regulator CDCA3 correlates with sensitivity to platinum agents in non-small cell lung cancer patients and cell lines. They show that interfering with CDCA3 degradation through CK2 inhibition enhances CDCA3 levels and increases sensitivity to platinum agents suggesting a therapeutic route.
- Published
- 2021
24. Mir‐34a and mir‐200c have an additive tumor‐suppressive effect on breast cancer cells and patient prognosis
- Author
-
Behzad Baradaran, Pascal H.G. Duijf, Vahid Khaze, Nicola Silvestris, Behzad Mansoori, Dariush Shanehbandi, Ahad Mokhtarzadeh, Afshin Derakhshani, Ali Mohammadi, Elham Baghbani, Mansoori, Behzad, Silvestris, Nicola, Mohammadi, Ali, Khaze, Vahid, Baghbani, Elham, Mokhtarzadeh, Ahad, Shanehbandi, Dariush, Derakhshani, Afshin, Duijf, Pascal HG, and Baradaran, Behzad
- Subjects
0301 basic medicine ,cancer stemness ,Cell cycle checkpoint ,Apoptosis ,CXCR4 ,Metastasis ,miR-200c ,Mice ,0302 clinical medicine ,Breast cancer ,Cell Movement ,Medicine ,Genetics (clinical) ,apoptosis ,Middle Aged ,Prognosis ,Progression-Free Survival ,Gene Expression Regulation, Neoplastic ,Matrix Metalloproteinase 9 ,cell cycle arrest ,030220 oncology & carcinogenesis ,Heterografts ,Female ,miR-34a ,HIF1-α ,Receptors, CXCR4 ,Epithelial-Mesenchymal Transition ,lcsh:QH426-470 ,MiR‐200c ,Breast Neoplasms ,HIF1‐α ,Malignancy ,Article ,Cell cycle arrest ,03 medical and health sciences ,breast cancer ,Downregulation and upregulation ,Cell Line, Tumor ,microRNA ,Genetics ,Animals ,Humans ,metastasis ,Cancer stemness ,Cell Proliferation ,business.industry ,Cancer ,MiR‐34a ,Hypoxia-Inducible Factor 1, alpha Subunit ,medicine.disease ,MicroRNAs ,lcsh:Genetics ,030104 developmental biology ,Cancer research ,business - Abstract
Breast cancer is the most common women’s malignancy in the world and, for subgroups ofpatients, treatment outcomes remain poor. Thus, more effective therapeutic strategies are urgently needed. MicroRNAs (miRNAs) have emerged as promising therapeutic tools and targets, as they play significant roles in regulating key cellular processes by suppressing gene expression. However, additive opportunities involving miRNAs have been underexplored. For example, both miR-34aand miR-200c individually suppress the development of different types of cancer, but the cellular effects of their combined actions remain unknown. Here, we show that miR-34a and miR-200c levels are reduced in breast tumors compared to adjacent normal tissues and that this additively predicts poor patient survival. In addition, in cell lines, miR-34a and miR-200c additively induce apoptosis and cell cycle arrest, while also inhibiting proliferation, invasion, migration, stemness andepithelial-to-mesenchymal transition (EMT). Mechanistically, both miRNA-34a and miR-200c directly target HIF1-α and subsequently downregulate VEGFR, MMP9 and CXCR4, although combined miRNA-34a and miR-200c delivery suppresses mouse xenograft tumor development as effectively as individual delivery. We establish a model, supported by in vitro and clinical data, which collectively suggest that the co-delivery of miR-34a and miR-200c represents a promising novel therapeuticstrategy for breast cancer patients. Breast cancer is the most common women’s malignancy in the world and, for subgroups of patients, treatment outcomes remain poor. Thus, more effective therapeutic strategies are urgently needed. MicroRNAs (miRNAs) have emerged as promising therapeutic tools and targets, as they play significant roles in regulating key cellular processes by suppressing gene expression. However, additive opportunities involving miRNAs have been underexplored. For example, both miR‐34a and miR‐200c individually suppress the development of different types of cancer, but the cellular effects of their combined actions remain unknown. Here, we show that miR‐34a and miR‐ 200c levels are reduced in breast tumors compared to adjacent normal tissues and that this additively predicts poor patient survival. In addition, in cell lines, miR‐34a and miR‐200c additively induce apoptosis and cell cycle arrest, while also inhibiting proliferation, invasion, migration, stemness and epithelial‐to‐mesenchymal transition (EMT). Mechanistically, both miRNA‐34a and miR‐ 200c directly target HIF1‐α and subsequently downregulate VEGFR, MMP9 and CXCR4, although combined miRNA‐34a and miR‐200c delivery suppresses mouse xenograft tumor development as effectively as individual delivery. We establish a model, supported by in vitro and clinical data, which collectively suggest that the co‐delivery of miR‐34a and miR‐200c represents a promising novel therapeutic strategy for breast cancer patients.
- Published
- 2021
- Full Text
- View/download PDF
25. Restoration of miR-330 expression suppresses lung cancer cell viability, proliferation, and migration
- Author
-
Grith Lykke Sørensen, Behzad Mansoori, Behrooz Shokouhi, Behzad Baradaran, Pascal H.G. Duijf, Leila Tebbi, Souzan Najafi, Ali Mohammadi, Elham Safarzadeh, Uffe Holmskov, Mohammadi, Ali, Mansoori, Behzad, Duijf, Pascal HG, Safarzadeh, Elham, Tebbi, Leila, Najafi, Souzan, Shokouhi, Behrooz, Sorensen, Grith L, Holmskov, Uffe, and Baradaran, Behzad
- Subjects
0301 basic medicine ,Lung Neoplasms ,Cell Survival ,Physiology ,Clinical Biochemistry ,Apoptosis ,Vimentin ,migration ,miR-330 ,miR‐330 ,03 medical and health sciences ,Chemokine receptor ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,microRNA ,Biomarkers, Tumor ,medicine ,Humans ,Genes, Tumor Suppressor ,RNA, Messenger ,Viability assay ,Lung cancer ,Cell Proliferation ,biology ,apoptosiscell cycle ,apoptosis ,Cell migration ,Cell Biology ,Cell cycle ,medicine.disease ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,lung cancer ,030104 developmental biology ,A549 Cells ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,cell cycle - Abstract
Lung cancer is one of the most common cancers and its incidence is rising around the world. Various studies suggest that miR-330 acts as a tumor-suppressor microRNA (miRNA) in different types of cancers, but precisely how has remained unclear. In this study, we investigate miR-330 expression in lung cancer patient samples, as well as in vitro, by studying how normalization of miR-330 expression affects lung cancer cellular phenotypes such as viability, apoptosis, proliferation, and migration. We establish that low miR-330 expression predicts poor lung cancer prognosis. Stable restoration of reduced miR-330 expression in lung cancer cells reduces cell viability, increases the fraction of apoptotic cells, causes G2/M cell cycle arrest, and inhibits cell migration. These findings are substantiated by increased mRNA and protein expression of markers for apoptosis via the intrinsic pathway, such as caspase 9, and decreased mRNA and protein expression of markers for cell migration, such as vimentin, C-X-C chemokine receptor type 4, and matrix metalloproteinase 9. We showed that reduced miR-330 expression predicts poor lung cancer survival and that stable restoration of miR-330 expression in lung cancer cells has a broad range of tumor-suppressive effects. This indicates that miR-330 is a promising candidate for miRNA replacement therapy for lung cancer patients. Refereed/Peer-reviewed
- Published
- 2021
- Full Text
- View/download PDF
26. The SWI/SNF subunit SMARCD3 regulates cell cycle progression and predicts survival outcome in ER+ breast cancer
- Author
-
Romain Tropée, Madeline Gough, Pascal H.G. Duijf, Cameron Snell, Eloise Dray, Bárbara de la Peña Avalos, Tropée, Romain, de la Peña Avalos, Bárbara, Gough, Madeline, Snell, Cameron, Duijf, Pascal HG, and Dray, Eloïse
- Subjects
0301 basic medicine ,Cancer Research ,Sucrose ,Cell cycle checkpoint ,SMARCD3 ,Breast Neoplasms ,Biology ,Chromatin remodeling ,Article ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,breast cancer ,medicine ,Endoreduplication ,Humans ,DNA damage repair ,Cell Cycle ,Cancer ,Cell cycle ,medicine.disease ,SWI/SNF ,Chromatin ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,cell cycle ,DNA Damage ,Transcription Factors - Abstract
Purpose: Chromatin remodeling plays an essential role in regulating transcriptional networks and timing of gene expression. Chromatin remodelers such as SWItch/Sucrose Non-Fermentable (SWI/SNF) harbor many protein components, with the catalytic subunit providing ATPase activity to displace histones along or from the DNA molecules, and associated subunits ensuring tissue specificity and transcriptional or co-transcriptional activities. Mutations in several of the SWI/SNF subunits have been linked to cancer. Here, we investigate between SMARCD3/Baf60c expression and hormone-positive (ER+) breast cancer. Methods: The level of SMARCD3 was detected by immunohistochemistry in breast cancer patient samples, and expression levels of SMARCD1, SMARCD2, and SMARCD3 were investigated using publicly available datasets from large cohorts of breast cancer patients. Using molecular biology and microscopy, we interrogated the cellular consequences of lower SMARCD3 expression. Results: Lower proliferation rates were observed in SMARCD3-depleted cells, which reflects a failure of the cell cycle progression and an increase in endoreplication. In the absence of SMARCD3, p21 accumulates in cells, but does not halt the cell cycle, and DNA damage accumulates and remains unrepaired. Conclusion: Taken together, our data begin to explain why ER+ breast cancer patients with low-SMARCD3 expressing tumors exhibit reduced survival rates compared to patients expressing normal or higher levels of SMARCD3. SMARCD3 might act as a tumor suppressor through regulation of cell cycle checkpoints and could be a reliable and specific breast cancer prognostic biomarker. Refereed/Peer-reviewed
- Published
- 2021
27. STAT3 Silencing and TLR7/8 Pathway Activation Repolarize and Suppress Myeloid-Derived Suppressor Cells From Breast Cancer Patients
- Author
-
Elham Safarzadeh, Ali Mohammadi, Behzad Mansoori, Pascal H. G. Duijf, Shahryar Hashemzadeh, Vahid Khaze, Tohid Kazemi, Afshin Derakhshani, Nicola Silvestris, Behzad Baradaran, Safarzadeh, Elham, Mohammadi, Ali, Mansoori, Behzad, Duijf, Pascal HG, Hashemzadeh, Shahryar, Khaze, Vahid, Kazemi, Tohid, Derakhshani, Afshin, Silvestris, Nicola, and Baradaran, Behzad
- Subjects
lcsh:Immunologic diseases. Allergy ,STAT3 Transcription Factor ,CD3 Complex ,medicine.medical_treatment ,CD3 ,T cell ,T-Lymphocytes ,Immunology ,Breast Neoplasms ,TLR7/8 ,Immunophenotyping ,STAT3 ,Breast cancer ,Immune system ,breast cancer ,Immune Tolerance ,Tumor Microenvironment ,Immunology and Allergy ,Medicine ,Humans ,Myeloid Cells ,Cell Proliferation ,Original Research ,Tumor microenvironment ,biology ,business.industry ,Myeloid-Derived Suppressor Cells ,Cell Differentiation ,Immunotherapy ,Transfection ,HLA-DR Antigens ,Middle Aged ,medicine.anatomical_structure ,Toll-Like Receptor 7 ,Myeloid-derived suppressor cells ,Toll-Like Receptor 8 ,Cancer cell ,Cancer research ,Myeloid-derived Suppressor Cell ,biology.protein ,Cytokines ,Female ,lcsh:RC581-607 ,business ,Signal Transduction - Abstract
Cancer cells escape immune destruction. From this perspective, myeloid-derived suppressor cells (MDSCs), which are immunosuppressive in various cancers including breast cancer (BC), are significant. However, the precise mechanisms are unknown. We isolated HLA-DR-CD33+ MDSCs and CD3+ T cells from BC patients’ peripheral blood and healthy donors through MACS and immunophenotyped by flow cytometry. Transfection of short-interfering RNAs and treatment with a TLR7/8 agonist altered pathway activities in vitro. Gene expression was analyzed using qRT-PCR, western blotting, and immunohistochemistry. Our findings showed an association between the progression of BC and increased levels of circulating HLA-DR-CD33+ MDSCs. These cells strongly suppress both autologous and analogous CD3+ T cell proliferation and enter the tumor microenvironment. We also identified increased STAT3 signaling and increased IDO and IL-10 expression in BC-derived MDSCs as immunosuppression mechanisms. Further, STAT3 inhibition and TLR7/8 pathway stimulation reduce the immunosuppressive activity of patient-derived MDSCs on T cells by inducing MDSC repolarization and differentiation into mature myeloid cells. This also alters the expression of critical cytokines and transcription factors in CD3+ T cells and, importantly, reduces breast cancer cells’ proliferation. Finally, while chemotherapy is able to significantly reduce circulating MDSCs’ level in patients with breast cancer, these MDSCs remained highly T cell-suppressive. We identified a novel molecular mechanism of MDSC-mediated immunosuppression. STAT3 inhibition and TLR7/8 pathway stimulation in MDSCs repolarize and suppress MDSCs from breast cancer patients. This offers new opportunities for BC immunotherapy. Cancer cells escape immune destruction. From this perspective, myeloid-derived suppressor cells (MDSCs), which are immunosuppressive in various cancers including breast cancer (BC), are significant. However, the precise mechanisms are unknown. We isolated HLA-DR-CD33+ MDSCs and CD3+ T cells from BC patients’ peripheral blood and healthy donors through MACS and immunophenotyped by flow cytometry. Transfection of short-interfering RNAs and treatment with a TLR7/8 agonist altered pathway activities in vitro. Gene expression was analyzed using qRT-PCR, western blotting, and immunohistochemistry. Our findings showed an association between the progression of BC and increased levels of circulating HLA-DR-CD33+ MDSCs. These cells strongly suppress both autologous and analogous CD3+ T cell proliferation and enter the tumor microenvironment. We also identified increased STAT3 signaling and increased IDO and IL-10 expression in BC-derived MDSCs as immunosuppression mechanisms. Further, STAT3 inhibition and TLR7/8 pathway stimulation reduce the immunosuppressive activity of patient-derived MDSCs on T cells by inducing MDSC repolarization and differentiation into mature myeloid cells. This also alters the expression of critical cytokines and transcription factors in CD3+ T cells and, importantly, reduces breast cancer cells’ proliferation. Finally, while chemotherapy is able to significantly reduce circulating MDSCs’ level in patients with breast cancer, these MDSCs remained highly T cell-suppressive. We identified a novel molecular mechanism of MDSC-mediated immunosuppression. STAT3 inhibition and TLR7/8 pathway stimulation in MDSCs repolarize and suppress MDSCs from breast cancer patients. This offers new opportunities for BC immunotherapy.
- Published
- 2020
- Full Text
- View/download PDF
28. Overexpression of HMGA2 in breast cancer promotes cell proliferation, migration, invasion and stemness
- Author
-
Behzad Baradaran, Henrik J. Ditzel, Pascal H.G. Duijf, Khalil Hajiasgharzadeh, Elmira Roshani, Morten F. Gjerstorff, Dariush Shanehbandi, Solmaz Shirjang, Souzan Najafi, Behzad Mansoori, Ali Mohammadi, Ahad Mokhtarzadeh, Tohid Kazemi, Mansoori, Behzad, Duijf, Pascal HG, Mohammadi, Ali, Najafi, Souzan, Roshani, Elmira, Shanehbandi, Dariush, Hajiasgharzadeh, Khalil, Shirjang, Solmaz, Ditzel, Henrik J, Kazemi, Tohid, Mokhtarzadeh, Ahad, Gjerstorff, Morten F, and Baradaran, Behzad
- Subjects
0301 basic medicine ,Cell cycle checkpoint ,HMGA2 ,proliferation ,Clinical Biochemistry ,03 medical and health sciences ,stemness ,0302 clinical medicine ,Breast cancer ,breast cancer ,Cancer stem cell ,Drug Discovery ,medicine ,CD133 ,Pharmacology ,biology ,Cell growth ,apoptosis ,EMT ,Cancer ,Cell cycle ,medicine.disease ,030104 developmental biology ,Apoptosis ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Molecular Medicine ,cell cycle - Abstract
Despite improved therapeutic strategies for early-stage breast cancer, the most common cancer type in women, relapse remains common and the underlying mechanisms for this progression remain poorly understood. To gain more insight, we studied the DNA-binding protein HMGA2 in breast cancer development and stemness. We demonstrated that HMGA2 is overexpressed in breast cancer tissues at the mRNA and protein levels (P value
- Published
- 2020
- Full Text
- View/download PDF
29. SASH1 is a prognostic indicator and potential therapeutic target in non-small cell lung cancer
- Author
-
Pascal H.G. Duijf, Gavin M. Wright, Shu-Dong Zhang, Emma Bolderson, Steven G. Gray, Joshua T. Burgess, Mark N. Adams, Derek J. Richard, Kenneth J. O'Byrne, Burgess, Joshua T, Bolderson, Emma, Adams, Mark N, Duijf, Pascal HG, Zhang, Shu‑Dong, Gray, Steven G, Wright, Gavin, Richard, Derek J, and O'Byrne, Kenneth J
- Subjects
0301 basic medicine ,Cell death ,Cell biology ,Lung Neoplasms ,Cancer therapy ,Cell ,lcsh:Medicine ,Apoptosis ,Treatment of lung cancer ,Article ,03 medical and health sciences ,Cell growth ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Medicine ,Humans ,RNA, Messenger ,SASH1 ,lcsh:Science ,Lung cancer ,Cancer ,Cell Proliferation ,tumor suppressor protein ,Cisplatin ,Multidisciplinary ,business.industry ,Tumor Suppressor Proteins ,lcsh:R ,Chloropyramine ,apoptosis ,mRNA expression ,medicine.disease ,Prognosis ,Gene Expression Regulation, Neoplastic ,non‑small cell lung cancer ,030104 developmental biology ,medicine.anatomical_structure ,A549 Cells ,030220 oncology & carcinogenesis ,Cancer research ,Adenocarcinoma ,lcsh:Q ,business ,medicine.drug - Abstract
SASH1 (SAM and SH3 domain-containing protein 1) is a tumor suppressor protein that has roles in key cellular processes including apoptosis and cellular proliferation. As these cellular processes are frequently disrupted in human tumours and little is known about the role of SASH1 in the pathogenesis of the disease, we analysed the prognostic value of SASH1 in non-small cell lung cancers using publicly available datasets. Here, we show that low SASH1 mRNA expression is associated with poor survival in adenocarcinoma. Supporting this, modulation of SASH1 levels in a panel of lung cancer cell lines mediated changes in cellular proliferation and sensitivity to cisplatin. The treatment of lung cancer cells with chloropyramine, a compound that increases SASH1 protein concentrations, reduced cellular proliferation and increased sensitivity to cisplatin in a SASH1-dependent manner. In summary, compounds that increase SASH1 protein levels could represent a novel approach to treat NSCLC and warrant further study.
- Published
- 2020
30. Association of sperm-associated antigen 5 and treatment response in patients with estrogen receptor-positive breast cancer
- Author
-
Andrew R. Green, Sunil R. Lakhani, Stephen Chan, Amy E. McCart Reed, Carlos Caldas, Graham Ball, Peter T. Simpson, Lynne Reid, Pulari U. Thangavelu, Alan Graham Pockley, Ian O. Ellis, Lorinc Pongor, Pascal H.G. Duijf, Jodi M. Saunus, Paul M. Moseley, Balazs Gyorffy, Tarek M. A. Abdel-Fatah, Abdel-Fatah, Tarek MA, Ball, Graham R, Thangavelu, Pulari U, Reid, Lynne E, McCart Reed, Amy E, Saunus, Jodi M, Duijf, Pascal HG, Simpson, Peter T, Lakhani, Sunil R, Pongor, Lorinc, Gyorffy, Balázs, Moseley, Paul M, Green, Andrew R, Pockley, Alan G, Caldas, Carlos, Ellis, Ian O, and Chan, Stephen YT
- Subjects
Oncology ,medicine.medical_specialty ,medicine.drug_class ,medicine.medical_treatment ,Estrogen receptor ,Antineoplastic Agents ,Breast Neoplasms ,Cell Cycle Proteins ,anthracycline ,Biomarkers, Pharmacological ,Breast cancer ,Estrogen Receptor Modulators ,Internal medicine ,medicine ,Humans ,Anthracyclines ,Progression-free survival ,education ,Neoadjuvant therapy ,SPAG5 protein ,Neoplasm Staging ,Original Investigation ,education.field_of_study ,business.industry ,Gene Expression Profiling ,Research ,Sperm associated antigen 5 ,Combination chemotherapy ,General Medicine ,Middle Aged ,medicine.disease ,Chemotherapy regimen ,Progression-Free Survival ,adjuvant chemotherapy ,Online Only ,Receptors, Estrogen ,Estrogen ,Chemotherapy, Adjuvant ,Drug Resistance, Neoplasm ,Female ,Estrogen Receptor Antagonists ,Drug Monitoring ,business ,estrogen receptor - Abstract
Key Points Question Are sperm-associated antigen 5 (SPAG5) transcript or protein expressions associated with treatment response in patients with estrogen receptor–positive breast cancer? Findings In this cohort study including 12 720 patients with estrogen receptor–positive breast cancer, SPAG5 transcript and SPAG5 protein overexpressions were associated with worse outcomes in patients who received endocrine therapy alone. Overexpressions of SPAG5 transcript or SPAG5 protein were associated with resistance to endocrine therapy but sensitivity to anthracycline-based combination chemotherapy, and downregulation of SPAG5 during the course of preoperative systemic therapies was associated with clinical benefit. Meaning These findings suggest that SPAG5 transcript or SPAG5 protein expression could be used as a clinical tool for selecting and monitoring response to neoadjuvant therapies and guide adjuvant therapy in estrogen receptor–positive breast cancer., This cohort study examines the association of sperm-associated antigen 5 (SPAG5) transcript and protein expression with treatment response in patients with estrogen receptor–positive breast cancer., Importance There is no proven test that can guide the optimal treatment, either endocrine therapy or chemotherapy, for estrogen receptor–positive breast cancer. Objective To investigate the associations of sperm-associated antigen 5 (SPAG5) transcript and SPAG5 protein expressions with treatment response in systemic therapy for estrogen receptor–positive breast cancer. Design, Settings, and Participants This retrospective cohort study included patients with estrogen receptor–positive breast cancer who received 5 years of adjuvant endocrine therapy with or without neoadjuvant anthracycline-based combination chemotherapy (NACT) derived from 11 cohorts from December 1, 1986, to November 28, 2019. The associations of SPAG5 transcript and SPAG5 protein expression with pathological complete response to NACT were evaluated, as was the association of SPAG5 mRNA expression with response to neoadjuvant endocrine therapy. The associations of distal relapse–free survival with SPAG5 transcript or SPAG5 protein expressions were analyzed. Data were analyzed from September 9, 2015, to November 28, 2019. Main Outcomes and Measures The primary outcomes were breast cancer–specific survival, distal relapse–free survival, pathological complete response, and clinical response. Outcomes were examined using Kaplan-Meier, multivariable logistic, and Cox regression models. Results This study included 12 720 women aged 24 to 78 years (mean [SD] age, 58.46 [12.45] years) with estrogen receptor–positive breast cancer, including 1073 women with SPAG5 transcript expression and 361 women with SPAG5 protein expression of locally advanced disease stage IIA through IIIC. Women with SPAG5 transcript and SPAG5 protein expressions achieved higher pathological complete response compared with those without SPAG5 transcript or SPAG5 protein expressions (transcript: odds ratio, 2.45 [95% CI, 1.71-3.51]; P
- Published
- 2020
31. miR-330 suppresses EMT and induces apoptosis by downregulating HMGA2 in human colorectal cancer
- Author
-
Uffe Holmskov, Vahid Khaze, Morten F. Gjerstorff, Behzad Baradaran, Behzad Mansoori, Pascal H.G. Duijf, Souzan Najafi, Solmaz Shirjang, Dariush Shanehbandi, Ali Mohammadi, Sanaz Naghizadeh, Mansoori, Behzad, Mohammadi, Ali, Naghizadeh, Sanaz, Gjerstorff, Morten, Shanehbandi, Dariush, Shirjang, Solmaz, Najafi, Souzan, Holmskov, Uffe, Khaze, Vahid, Duijf, Pascal HG, and Baradaran, Behzad
- Subjects
0301 basic medicine ,STAT3 Transcription Factor ,Vascular Endothelial Growth Factor A ,Cell signaling ,Epithelial-Mesenchymal Transition ,HMGA2 ,Physiology ,Clinical Biochemistry ,Apoptosis ,colorectal cancer ,miR-330 ,Snail-1 ,miR‐330 ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Downregulation and upregulation ,Antigens, CD ,Cell Movement ,Cell Line, Tumor ,microRNA ,Gene expression ,Humans ,Genes, Tumor Suppressor ,Snail‐1 ,Smad3 Protein ,Protein kinase B ,Cell Proliferation ,biology ,HMGA2 Protein ,apoptosis ,Cell Biology ,Cadherins ,HCT116 Cells ,Vascular endothelial growth factor ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Snail Family Transcription Factors ,Colorectal Neoplasms ,Proto-Oncogene Proteins c-akt ,Transforming growth factor ,Signal Transduction ,Smad3 - Abstract
MicroRNAs (miRNAs) are important molecular regulatorsof cellular signaling and behavior. They alter gene expression by targeting messenger RNAs, including those encoding transcriptional regulators, such as HMGA2. While HMGA2 is oncogenic in various tumors, miRNAs may be oncogenic or tumor suppressive. Here, we investigate the expression of HMGA2 and the miRNA miR-330 in a patient with colorectal cancer (CRC) samples and their effects on oncogenic cellular phenotypes. We found that HMGA2 expression is increased and miR-330 expression is decreased in CRCs and each predicts poor long-term patient survival. Stably increased miR-330 expression in human colorectal cancer cells (HCT116) and SW480 CRC cell lines downregulate the oncogenic expression of HMGA2, a predicted miR-330 target. Additionally, this promotes apoptosis and decreases cell migration and viability. Consistently, it also decreases protein-level expression of markers for epithelial-to-mesenchymal-transition (Snail-1, E-cadherin, and Vascular endothelial growth factor receptors) and transforming growth factor β signaling (SMAD3), as well as phospho- Protein kinase B (AKT) and phospho-STAT3 levels. We conclude that miR-330 acts as a tumor suppressor miRNA in CRC by suppressing HMGA2 expression and reducing cell survival, proliferation, and migration. Thus, we identify miR-330 as a promising candidate for miRNA replacement therapy for patients with CRC. Refereed/Peer-reviewed
- Published
- 2020
- Full Text
- View/download PDF
32. Complexities of pharmacogenomic interactions in cancer
- Author
-
Pascal H.G. Duijf, Kum Kum Khanna, Khanna, Kum Kum, and Duijf, Pascal HG
- Subjects
0301 basic medicine ,Drug ,Cancer Research ,media_common.quotation_subject ,precision medicine ,drug response ,Computational biology ,Drug resistance ,03 medical and health sciences ,0302 clinical medicine ,Drug response ,Medicine ,cancer ,drug sensitivity ,media_common ,Cancer ,pharmacogenomics ,drug resistance ,business.industry ,Precision medicine ,medicine.disease ,genomic instability ,Author's Views ,030104 developmental biology ,030220 oncology & carcinogenesis ,Pharmacogenomics ,Molecular Medicine ,Cancer development ,business - Abstract
Genetic and genomic alterations drive cancer development. However, they may also constitute vulnerabilities, including increased drug sensitivity, which could be harnessed for precision medicine purposes. We discuss the highly complex pharmacogenomic interactions that are beginning to be disentangled and hurdles that may need to be overcome before cancer patients could benefit. Refereed/Peer-reviewed
- Published
- 2020
33. Circulating myeloid‐derived suppressor cells: An independent prognostic factor in patients with breast cancer
- Author
-
Shahryar Hashemzadeh, Mehdi Yousefi, Ali Mohammadi, Pascal H.G. Duijf, Vahid Khaze, Behzad Baradaran, Farhad Babaie, Hamed Mohammadi, Milad Asadi, Behzad Mansoori, Elham Safarzadeh, Tohid Kazemi, Safarzadeh, Elham, Shahryar, Hashemzadeh, Duijf, Pascal HG, Mansoori, Behzad, Khaze, Vahid, Mohammadi, Ali, Kazemi, Tohid, Yousefi, Mehdi, Asadi, Milad, Mohammadi, Hamed, Babaie, Farhad, and Baradaran, Behzad
- Subjects
Adult ,0301 basic medicine ,Myeloid ,Physiology ,T-Lymphocytes ,CD3 ,T cell ,Sialic Acid Binding Ig-like Lectin 3 ,Clinical Biochemistry ,CD33 ,Breast Neoplasms ,Lymphocyte Activation ,Peripheral blood mononuclear cell ,immunoediting ,03 medical and health sciences ,breast cancer ,immunosuppressive ,0302 clinical medicine ,Immune system ,Tumor Microenvironment ,metastasis ,tumor microenvironment ,Humans ,Medicine ,Cells, Cultured ,Aged ,Cell Proliferation ,Neoplasm Staging ,Tumor microenvironment ,biology ,business.industry ,Myeloid-Derived Suppressor Cells ,HLA-DR Antigens ,Cell Biology ,Middle Aged ,Coculture Techniques ,Phenotype ,030104 developmental biology ,medicine.anatomical_structure ,Case-Control Studies ,Lymphatic Metastasis ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Myeloid-derived Suppressor Cell ,Female ,Tumor Escape ,business - Abstract
Evading immune destruction is a hallmark of cancer. Myeloid-derived suppressor cells (MDSCs), a heterogeneous population of myeloid immune cells, are thought to foster the establishment of an immunosuppressive tumor microenvironment, but it remains unclear how. This study aims to determine the levels of circulating MDSCs and their subpopulations and test their immunosuppressive functions in patients with breast cancer (BC). We analyzed the fractions of MDSCs in freshly isolated peripheral blood mononuclear cells of patients with BC and healthy donors using flow cytometry. Circulating MDSCs were further phenotyped using fluorescently labeled antihuman monoclonal antibodies. Coculture experiments revealed the effects of MDSCs on CD3 + T cell response. Moreover, we correlated circulating MDSC levels with clinicopathological features of patients with BC. We show that the fraction of HLA-DR – CD33 + MDSCs in peripheral blood is about 10-fold higher in patients with BC than in healthy control individuals. The levels of all MDSC subpopulations, including monocytic and granulocytic MDSCs, are significantly elevated. Coculture experiments of purified HLA-DR – CD33 + MDSCs and CD3 + T cells demonstrate that T cell proliferation is more effectively inhibited by BC patient-derived MDSCs than by healthy control MDSCs. Moreover, increased circulating MDSC levels robustly associate with advanced BC stage and positive lymph node status. By being more abundant and more effective T cell suppressors, BC patient-derived circulating MDSCs exert a dual immunosuppressive effect. Our findings pave the way to develop novel diagnostic and immunotherapeutic strategies, aimed at detecting and inhibiting MDSCs in patients with BC. Refereed/Peer-reviewed
- Published
- 2018
- Full Text
- View/download PDF
34. Elevating CDCA3 Levels Enhances Tyrosine Kinase Inhibitor Sensitivity in TKI-Resistant EGFR Mutant Non-Small-Cell Lung Cancer
- Author
-
Yaowu He, Csilla Hasovits, Amanda L. Hudson, Rozelle Harvie, Viive M. Howell, Katherine B. Sahin, Priyakshi Kalita-de Croft, Christopher Molloy, Sunil R. Lakhani, Sarah A. Hayes, Hannah Kamitakahara, Tashbib Khan, Genevieve P. Ferguson, Derek J. Richard, Kenneth J. O'Byrne, Esha T. Shah, Emma Bolderson, Mark N. Adams, Emily Colvin, Pascal H.G. Duijf, John D. Hooper, Sahin, Katherine B, Shah, Esha T, Ferguson, Genevieve P, Molloy, Christopher, Duijf, Pascal HG, and Adams, Mark N
- Subjects
Cancer Research ,medicine.drug_class ,tyrosine kinase inhibitor (TKI) ,non-small-cell lung cancer (NSCLC) ,non-small cell lung cancer (NSCLC) ,Article ,Receptor tyrosine kinase ,Tyrosine-kinase inhibitor ,T790M ,medicine ,Epidermal growth factor receptor ,Lung cancer ,RC254-282 ,biology ,business.industry ,acquired resistance ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,epidermal growth factor receptor (EGFR) ,respiratory tract diseases ,cell division cycle-associated protein 3 (CDCA3) ,Oncology ,biology.protein ,Cancer research ,biomarker ,Casein kinase 2 ,business ,Tyrosine kinase - Abstract
Simple Summary Resistance to tyrosine kinase inhibitors (TKIs) that target common non-small-cell lung cancer mutations within the epidermal growth factor receptor (EGFR) is a primary clinical issue. The aim of our study was to determine whether the protein cell division cycle-associated protein 3 (CDCA3) might be a biomarker for TKI response in EGFR mutant lung cancer. Our previous work has demonstrated that CDCA3 is a marker of chemotherapy sensitivity in lung cancer. We provide evidence that CDCA3 levels are increased in EGFR mutant lung cancer and these levels are associated with sensitivity to TKIs. In addition, increasing the levels of CDCA3 enhances TKI sensitivity in models of TKI-resistant EGFR mutant lung cancer. Our findings propose that strategies to upregulate CDCA3 levels might improve TKI response in EGFR mutant lung cancer. Abstract Tyrosine kinase inhibitors (TKIs) are the first-line therapy for non-small-cell lung cancers (NSCLC) that harbour sensitising mutations within the epidermal growth factor receptor (EGFR). However, resistance remains a key issue, with tumour relapse likely to occur. We have previously identified that cell division cycle-associated protein 3 (CDCA3) is elevated in adenocarcinoma (LUAD) and correlates with sensitivity to platinum-based chemotherapy. Herein, we explored whether CDCA3 levels were associated with EGFR mutant LUAD and TKI response. We demonstrate that in a small-cohort tissue microarray and in vitro LUAD cell line panel, CDCA3 protein levels are elevated in EGFR mutant NSCLC as a result of increased protein stability downstream of receptor tyrosine kinase signalling. Here, CDCA3 protein levels correlated with TKI potency, whereby CDCA3high EGFR mutant NSCLC cells were most sensitive. Consistently, ectopic overexpression or inhibition of casein kinase 2 using CX-4945, which pharmacologically prevents CDCA3 degradation, upregulated CDCA3 levels and the response of T790M(+) H1975 cells and two models of acquired resistance to TKIs. Accordingly, it is possible that strategies to upregulate CDCA3 levels, particularly in CDCA3low tumours or upon the emergence of therapy resistance, might improve the response to EGFR TKIs and benefit patients.
- Published
- 2021
- Full Text
- View/download PDF
35. The synergy between miR-486–5p and tamoxifen causes profound cell death of tamoxifen-resistant breast cancer cells
- Author
-
William C. Cho, Elmira Roshani, Pascal H.G. Duijf, Behnaz Mansoori, Pouria Savadi, Behzad Mansoori, Afsaneh Nazari, Ahad Mokhtarzadeh, Ali Mohammadi, Haleh AsadollahSeraj, Behzad Baradaran, Souzan Najafi, Mansoori, Behzad, Najafi, Souzan, Mohammadi, Ali, AsadollahSeraj, Haleh, Savadi, Pouria, Mansoori, Behnaz, Nazari, Afsaneh, Mokhtarzadeh, Ahad, Roshani, Elmira, Duijf, Pascal HG, Cho, William Chi-Shing, and Baradaran, Behzad
- Subjects
HMGA1 ,Programmed cell death ,Antineoplastic Agents, Hormonal ,MiR-486–5p ,drug-resistant ,Cell ,Population ,Estrogen receptor ,Breast Neoplasms ,RM1-950 ,Breast cancer ,breast cancer ,Humans ,Medicine ,skin and connective tissue diseases ,education ,Drug-resistant ,Pharmacology ,education.field_of_study ,Cell Death ,Dose-Response Relationship, Drug ,tamoxifen ,business.industry ,Cancer ,Cell migration ,General Medicine ,medicine.disease ,MicroRNAs ,Tamoxifen ,HEK293 Cells ,medicine.anatomical_structure ,Drug Resistance, Neoplasm ,MCF-7 Cells ,Cancer research ,Female ,Therapeutics. Pharmacology ,business ,medicine.drug - Abstract
Breast cancer (BC) is the most common type of malignancy in women. A subset of breast cancers show resistance to endocrine-based therapies. The estrogen receptor (ER) plays a critical role in developing hormone-dependent BC. Loss of ER contributes to resistance to tamoxifen therapy and may contribute to mortality. Thus, it is crucial to overcome this problem. Here, using luciferase reporter assays, qRT-PCR, and Western blot analyses, we demonstrate that the microRNA miR-486–5p targets HMGA1 mRNA, decreasing its mRNA and protein levels in ER-positive (ER+) BC cells. Consistently, miR-486–5p is significantly downregulated, whereas HMGA1 is considerably upregulated in ER+ BC samples. Remarkably, while both miR-486–5p and tamoxifen individually cause G2/M cell cycle arrest, combination treatment synergistically causes profound cell death, specifically in tamoxifen-resistant ER+ cells but not in tamoxifen-sensitive ER+ cells. Combined treatment with miR-486–5p and tamoxifen also additively reduces cell migration, invasion, colony formation, mammary spheroid formation and a CD24-CD44+ cell population, representing decreased cancer stemness. However, these phenomena are independent of the tamoxifen responsiveness of the ER+ BC cells. Thus, miR-486–5p and tamoxifen exhibit additive and synergistic tumor-suppressive effects, most importantly causing profound cell death specifically in tamoxifen-resistant BC cells. Therefore, our work suggests that combining miR-486–5p replacement therapy with tamoxifen treatment is a promising strategy to treat endocrine therapy-resistant BC. Breast cancer (BC) is the most common type of malignancy in women. A subset of breast cancers show resistance to endocrine-based therapies. The estrogen receptor (ER) plays a critical role in developing hormone-dependent BC. Loss of ER contributes to resistance to tamoxifen therapy and may contribute to mortality. Thus, it is crucial to overcome this problem. Here, using luciferase reporter assays, qRT-PCR, and Western blot analyses, we demonstrate that the microRNA miR-486–5p targets HMGA1 mRNA, decreasing its mRNA and protein levels in ER-positive (ER+) BC cells. Consistently, miR-486–5p is significantly downregulated, whereas HMGA1 is considerably upregulated in ER+ BC samples. Remarkably, while both miR-486–5p and tamoxifen individually cause G2/M cell cycle arrest, combination treatment synergistically causes profound cell death, specifically in tamoxifen-resistant ER+ cells but not in tamoxifen-sensitive ER+ cells. Combined treatment with miR-486–5p and tamoxifen also additively reduces cell migration, invasion, colony formation, mammary spheroid formation and a CD24-CD44+ cell population, representing decreased cancer stemness. However, these phenomena are independent of the tamoxifen responsiveness of the ER+ BC cells. Thus, miR-486–5p and tamoxifen exhibit additive and synergistic tumor-suppressive effects, most importantly causing profound cell death specifically in tamoxifen-resistant BC cells. Therefore, our work suggests that combining miR-486–5p replacement therapy with tamoxifen treatment is a promising strategy to treat endocrine therapy-resistant BC.
- Published
- 2021
- Full Text
- View/download PDF
36. MiR-142-3p targets HMGA2 and suppresses breast cancer malignancy
- Author
-
Henrik J. Ditzel, Behzad Baradaran, Elham Safarzadeh, William C. Cho, Morten F. Gjerstorff, Behzad Mansoori, Pascal H.G. Duijf, Ali Mohammadi, Mansoori, Behzad, Duijf, Pascal HG, Mohammadi, Ali, Safarzadeh, Elham, Ditzel, Henrik J, Gjerstorff, Morten F, Cho, William Chi-Shing, and Baradaran, Behzad
- Subjects
Untranslated region ,HMGA2 ,miR-142-3p ,Breast Neoplasms ,Apoptosis ,Breast Neoplasms/genetics ,Malignancy ,General Biochemistry, Genetics and Molecular Biology ,STAT3 ,Breast cancer ,Cell Movement ,Cancer stem cell ,Gene expression ,microRNA ,Biomarkers, Tumor ,Tumor Cells, Cultured ,medicine ,Humans ,General Pharmacology, Toxicology and Pharmaceutics ,Cell Proliferation ,biology ,Cancer stem cells ,business.industry ,AKT ,HMGA2 Protein ,Cell Cycle ,General Medicine ,Cell cycle ,Prognosis ,medicine.disease ,HMGA2 Protein/genetics ,Gene Expression Regulation, Neoplastic ,Survival Rate ,MicroRNAs ,ERK ,MicroRNAs/genetics ,biology.protein ,Cancer research ,Female ,Biomarkers, Tumor/genetics ,business - Abstract
MicroRNAs (miRNAs) have the ability to regulate gene expression programs in cells. Hence, altered expression of miRNAs significantly contributes to breast cancer development and progression. Here, we demonstrate that the miRNA miR-142-3p directly targets the 3′ untranslated region of HMGA2, which encodes an onco-embryonic protein that is overexpressed in most cancers, including breast cancer. Down regulation of miR-142-3p predicting poor patient survival in grade 3 breast cancer (P-value = 0.045). MiR-142-3p downregulates HMGA2 mRNA and protein levels. Higher miR-142-3p and lower HMGA2 expressed are found in breast cancer versus normal breast tissue (P-value
- Published
- 2021
- Full Text
- View/download PDF
37. Silencing of BACH1 inhibits invasion and migration of prostate cancer cells by altering metastasis-related gene expression
- Author
-
Shima Salehi, Vahid Khaze Shahgoli, Tohid Kazemi, Neda Shajari, Sadaf Davudian, Ali Mohammadi, Behzad Mansoori, Behzad Baradaran, Dariush Shanehbandi, Pascal H.G. Duijf, Shajari, Neda, Davudian, Sadaf, Kazemi, Tohid, Mansoori, Behzad, Salehi, Shima, Shahgoli, Vahid Khaze, Shanehbandi, Dariush, Mohammadi, Ali, Duijf, Pascal HG, and Baradaran, Behzad
- Subjects
Male ,0301 basic medicine ,Biomedical Engineering ,Pharmaceutical Science ,Medicine (miscellaneous) ,Biology ,migration ,Metastasis ,gene silencing ,03 medical and health sciences ,Prostate cancer ,Engineering ,RNA interference. siRNA ,0302 clinical medicine ,DU145 ,Cell Movement ,Prostate ,LNCaP ,medicine ,Humans ,Gene silencing ,Neoplasm Invasiveness ,Gene Silencing ,RNA, Messenger ,Neoplasm Metastasis ,RNA, Small Interfering ,lET-7 ,Engineering, Biomedical ,Materials Science, Biomaterials ,Prostatic Neoplasms ,Cancer ,General Medicine ,Chromoplexy ,invasion ,medicine.disease ,Molecular biology ,Gene Expression Regulation, Neoplastic ,Basic-Leucine Zipper Transcription Factors ,030104 developmental biology ,medicine.anatomical_structure ,Biotechnology & Applied Microbiology ,030220 oncology & carcinogenesis ,Cancer research ,Biotechnology - Abstract
Background: Cancer lethality is mainly caused by metastasis. Therefore, understanding the nature of the genes involved in this process has become a priority. BACH1, a basic leucine zipper transcription factor, has been shown to transcriptionally regulate expression of a range of genes that are associated with breast cancer metastasis. However, the exact role and the underlying molecular mechanism of BACH1 in prostate cancer remain unclear. This study aims to explore the expression of BACH1 in prostate cancer tissues and the effect of BACH1 suppression on prostate cancer cell behavior. Materials and methods: In this study, we used quantitative real-time PCR (qRT-PCR) to measure BACH1 expression in prostate adenocarcinoma tissues and two metastasis-derived prostate cancer cell lines, DU145 and LNCaP. We also used immunohistochemical (IHC) staining to measure BACH1 protein expression in prostate adenocarcinoma and matched normal tissue samples. In the following BACH1 expression was silenced in DU145 cells using siRNA as well. Knockdown was confirmed by qRT-PCR and Western blotting. The cytotoxic effects of BACH1-siRNA on DU145 cells were determined using an MTT assay. The migration and invasive capacity of DU145 cells were examined by scratch wound healing assay and matrigel invasion assay, respectively. We also used qRT-PCR to study the effect of BACH1 silencing on the expression levels of metastasis-related genes. Results:We find that the expression of BACH1 mRNA and protein in prostate cancer tissues is significantly higher than in matched normal prostate tissues (p
- Published
- 2017
- Full Text
- View/download PDF
38. Defining COMMD4 as an anti-cancer therapeutic target and prognostic factor in non-small cell lung cancer
- Author
-
Amila Suraweera, Alex Duff, Mark N. Adams, Christian Jekimovs, Pascal H. G. Duijf, Cheng Liu, Matthew McTaggart, Sam Beard, Kenneth J. O’Byrne, Derek J. Richard, Suraweera, Amila, Duff, Alex, Adams, Mark N, Jekimovs, Christian, Duijf, Pascal HG, Liu, Cheng, McTaggart, Matthew, Beard, Sam, O'Byrne, Kenneth J, and Richard, Derek J
- Subjects
Male ,tumor ,Cancer Research ,Lung Neoplasms ,Cell Survival ,carcinoma ,Article ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Biomarkers, Tumor ,Humans ,neoplasms ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Neoplasm Staging ,0303 health sciences ,camptothecin ,biomarkers ,Computational Biology ,Correction ,Prognosis ,Survival Analysis ,respiratory tract diseases ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,non-small-cell lung cancer ,Oncology ,Tissue Array Analysis ,030220 oncology & carcinogenesis ,Female ,Non-small-cell lung cancer ,copper metabolism protein - Abstract
Background Non-small cell lung cancers (NSCLC) account for 85–90% of all lung cancers. As drug resistance critically impairs chemotherapy effectiveness, there is great need to identify new therapeutic targets. The aims of this study were to investigate the prognostic and therapeutic potential of the copper-metabolism-domain-protein, COMMD4, in NSCLC. Methods The expression of COMMD4 in NSCLC was investigated using bioinformatic analysis, immunoblotting of immortalised human bronchial epithelial (HBEC) and NSCLC cell lines, qRT-PCR and immunohistochemistry of tissue microarrays. COMMD4 function was additionally investigated in HBEC and NSCLC cells depleted of COMMD4, using small interfering RNA sequences. Results Bioinformatic analysis and in vitro analysis of COMMD4 transcripts showed that COMMD4 levels were upregulated in NSCLC and elevated COMMD4 was associated with poor prognosis in adenocarcinoma (ADC). Immunoblotting demonstrated that COMMD4 expression was upregulated in NSCLC cells and siRNA-depletion of COMMD4, decreased cell proliferation and reduced cell viability. Cell death was further enhanced after exposure to DNA damaging agents. COMMD4 depletion caused NSCLC cells to undergo mitotic catastrophe and apoptosis. Conclusions Our data indicate that COMMD4 may function as a prognostic factor in ADC NSCLC. Additionally, COMMD4 is a potential therapeutic target for NSCLC, as its depletion induces cancer cell death.
- Published
- 2019
39. HMGA2 and Bach-1 cooperate to promote breast cancer cell malignancy
- Author
-
Pascal H.G. Duijf, Neda Shajari, Zahra Asadzadeh, Tohid Kazemi, Behzad Baradaran, Fereydoon Abedi Gaballu, Solmaz Shirjang, Morten F. Gjerstorff, Behzad Mansoori, Ali Mohammadi, Mahsa Minouei, Mansoori, Behzad, Mohammadi, Ali, Asadzadeh, Zahra, Shirjang, Solmaz, Minouei, Mahsa, Abedi Gaballu, Fereydoon, Shajari, Neda, Kazemi, Tohid, Gjerstorff, Morten F, Duijf, Pascal HG, and Baradaran, Behzad
- Subjects
0301 basic medicine ,cancer stem cell ,Epithelial-Mesenchymal Transition ,HMGA2 ,Physiology ,Clinical Biochemistry ,Down-Regulation ,Apoptosis ,Breast Neoplasms ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,breast cancer ,Cancer stem cell ,Cell Movement ,Cell Line, Tumor ,medicine ,Humans ,Transcription factor ,Cell Proliferation ,Gene knockdown ,biology ,Cell growth ,bach1 ,HMGA2 Protein ,apoptosis ,EMT ,Cell Biology ,Cell cycle ,Middle Aged ,medicine.disease ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Basic-Leucine Zipper Transcription Factors ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Disease Progression ,Female - Abstract
During breast cancer progression, tumor cells acquire multiple malignant features. The transcription factors and cell cycle regulators high mobility group A2 (HMGA2)and BTB and CNC homology 1 (Bach‐1) are overexpressed in several cancers, but the mechanistic understanding of how HMGA2 and Bach‐1 promote cancer development has been limited. We found that HMGA2 and Bach‐1 are overexpressed in breast cancer tissues and their expression correlates positively in tumors but not in normal tissues. Individual HMGA2 or Bach‐1 knockdown downregulates expression of both proteins, suggesting a mutual stabilizing effect between the two proteins. Importantly, combined HMGA2 and Bach‐1 knockdown additively decrease cell proliferation, migration, epithelial‐to‐mesenchymal transition, and colony formation, while promoting apoptotic cell death via upregulation of caspase‐3 and caspase‐9.First the first time, we show that HMGA2 and Bach‐1 overexpression in tumors correlate positively and that the proteins cooperatively suppress a broad range of malignant cellular properties, such as proliferation, migration, clonogenicity, and evasion of apoptotic cell death. Thus, our observations suggest that combined targeting of HMGA2 and Bach1 may be an effective therapeutic strategy to treat breast cancer Refereed/Peer-reviewed
- Published
- 2019
- Full Text
- View/download PDF
40. Large expert-curated database for benchmarking document similarity detection in biomedical literature search
- Author
-
Brown, Peter, Zhou, Yaoqi, Li, Jiuyong, Lim, Renly, You, Yuyi, Li, Ying, Le, Thuc D, Cao, Zehong, Duijf, Pascal HG, and RELISH Consortium
- Abstract
Document recommendation systems for locating relevant literature have mostly relied on methods developed a decade ago. This is largely due to the lack of a large offline gold-standard benchmark of relevant documents that cover a variety of research fields such that newly developed literature search techniques can be compared, improved and translated into practice. To overcome this bottleneck, we have established the RElevant LIterature SearcH consortium consisting of more than 1500 scientists from 84 countries, who have collectively annotated the relevance of over 180 000 PubMed-listed articles with regard to their respective seed (input) article/s. The majority of annotations were contributed by highly experienced, original authors of the seed articles. The collected data cover 76% of all unique PubMed Medical Subject Headings descriptors.No systematic biases were observed across different experience levels, research fields or time spent on annotations. More importantly, annotations of the same document pairs contributed by different scientists were highly concordant. We further show that the three representative baseline methods used to generate recommended articles for evaluation (Okapi Best Matching 25, Term Frequency–Inverse Document Frequency and PubMed Related Articles) had similar overall performances. Additionally, we found that these methods each tend to produce distinct collections of recommended articles,suggesting that a hybrid method may be required to completely capture all relevant articles. The established database server located at https://relishdb.ict.griffith.edu.au is freely available for the downloading of annotation data and the blind testing of new methods. We expect that this benchmark will be useful for stimulating the development of new powerful techniques for title and title/abstract-based search engines for relevant articles in biomedical research. Refereed/Peer-reviewed
- Published
- 2019
41. A functional genetic screen identifies the Mediator complex as essential for SSX2-induced senescence
- Author
-
Sofie N. Bennedsen, Henrik J. Ditzel, Torben A Kruse, Nicolas Alcaraz, Morten F. Gjerstorff, Nadine Heidi Brückmann, Martin Jakob Larsen, Mads Thomassen, Pascal H.G. Duijf, Mikkel G. Terp, Christina Bøg Pedersen, Brückmann, Nadine H, Bennedsen, Sofie N, Duijf, Pascal HG, Terp, Mikkel G, Thomassen, Mads, Larsen, Martin, Pedersen, Christina B, Kruse, Torben, Alcaraz, Nicolas, Ditzel, Henrik J, and Gjerstorff, Morten F
- Subjects
Transcriptional Activation ,Senescence ,Cancer Research ,Transcription, Genetic ,Immunology ,Mediator ,Biology ,medicine.disease_cause ,Article ,MED1 ,Mediator Complex Subunit 1 ,Sarcoma, Synovial ,Cellular and Molecular Neuroscience ,Downregulation and upregulation ,Cell Line, Tumor ,medicine ,Humans ,cellular senescence ,lcsh:QH573-671 ,Melanoma ,Cellular Senescence ,Cell Nucleus ,Gene knockdown ,Mediator Complex ,lcsh:Cytology ,cell nucleus ,Oncogenes ,Cell Biology ,Neoplasm Proteins ,Chromatin ,Cell biology ,Gene Expression Regulation, Neoplastic ,Repressor Proteins ,SSX2 ,Carcinogenesis ,Protein Kinases ,Transcription Factors ,Genetic screen - Abstract
The senescence response to oncogenes is believed to be a barrier to oncogenic transformation in premalignant lesions, and describing the mechanisms by which tumor cells evade this response is important for early diagnosis and treatment. The male germ cell-associated protein SSX2 is ectopically expressed in many types of cancer and is functionally involved in regulating chromatin structure and supporting cell proliferation. Similar to many well-characterized oncogenes, SSX2 has the ability to induce senescence in cells. In this study, we performed a functional genetic screen to identify proteins implicated in SSX2-induced senescence and identified several subunits of the Mediator complex, which is central in regulating RNA polymerase-mediated transcription. Further experiments showed that reduced levels of MED1, MED4, and MED14 perturbed the development of senescence in SSX2-expressing cells. In contrast, knockdown of MED1 did not prevent development of B-Raf- and Epirubicin-induced senescence, suggesting that Mediator may be specifically linked to the cellular functions of SSX2 that may lead to development of senescence or be central in a SSX2-specific senescence response. Indeed, immunostaining of melanoma tumors, which often express SSX proteins, exhibited altered levels of MED1 compared to benign nevi. Similarly, RNA-seq analysis suggested that MED1, MED4, and MED14 were downregulated in some tumors, while upregulated in others. In conclusion, our study reveals the Mediator complex as essential for SSX2-induced senescence and suggests that changes in Mediator activity could be instrumental for tumorigenesis.
- Published
- 2019
- Full Text
- View/download PDF
42. Downregulation of miR-146a promotes cell migration in Helicobacter pylori-negative gastric cancer
- Author
-
Behzad Mansoori, Mehdi Yousefi, Pascal H.G. Duijf, Dariush Shanehbandi, Navid Shomali, Naghmeh Shirafkan, Elham Baghbani, Behzad Baradaran, Zohreh Babaloo, Mehri Ghasabi, Ali Mohammadi, Milad Asadi, Shomali, Navid, Shirafkan, Naghmeh, Duijf, Pascal HG, Ghasabi, Mehri, Babaloo, Zohreh, Yousefi, Mehdi, Mansoori, Behzad, Asadi, Milad, Shanehbandi, Dariush, Baghbani, Elham, Mohammadi, Ali, and Baradaran, Behzad
- Subjects
Male ,0301 basic medicine ,Carcinogenesis ,Cell Survival ,Down-Regulation ,Apoptosis ,Helicobacter pylori-negative patients ,Biochemistry ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Cell Movement ,Stomach Neoplasms ,Cell Line, Tumor ,Gene expression ,microRNA ,medicine ,Humans ,metastasis ,Neoplasm Metastasis ,Molecular Biology ,Aged ,Cell Proliferation ,Helicobacter pylori ,biology ,Oncogene ,miR‐146a ,gastric cancer ,apoptosis ,Cancer ,Cell migration ,Cell Biology ,Middle Aged ,biology.organism_classification ,medicine.disease ,Survival Analysis ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Female - Abstract
microRNAs (miRs) are short noncoding RNAs that post‐transcriptionally suppress gene expression. miR‐146a acts as an oncogene or a tumor suppressor in various cancers, including gastric cancer, but it is unclear what determines whether miR‐146a is oncogenic or tumor suppressive and the molecular mechanisms are still largely unknown. The aim of this study was to investigate the role of miR‐146a in gastric cancer, by focusing on its expression in patients who were negative for Helicobacter pylori and its reduced and increased expression effect in vitro. Twenty gastric cancer patients who were negative for H. pylori infection were selected and the expression levels of miRNA‐146a in these gastric tumors, in their matched normal gastric tissues and in gastric cancer cell lines with varying tumorigenic potential was measured. Further, the impact of increased and decreased miR-146a expression levels on the expression of predicted target genes, cell migration, viability, proliferation, and apoptosis was examined, respectively. Our results for the first time indicated that miR-146a is downregulated in H. pylori-negative gastric cancers and suggests that H. pylori infection determines whether miR-146a acts as an oncogene or tumor suppressor. The level of miR-146a expression inversely correlates with the tumorigenicity of three gastric cancer cell lines and low miR-146a expression predicts poor recurrence-free survival. It was also found that miR-146a reduces the expression levels of the prometastatic genes and suppresses MKN-45 cell migration. Functional studies showed that miR-146a acts as a tumor suppressor miR and identifies miR-146a as a candidate for antimetastatic miRNA replacement therapy for gastric cancer patients. Refereed/Peer-reviewed
- Published
- 2019
43. Multi-omics characterization of the spontaneous mesenchymal-epithelial transition in the PMC42 breast cancer cell lines
- Author
-
Tony Blick, Erik W. Thompson, Sugandha Bhatia, Pascal H.G. Duijf, James Monkman, Shivashankar H. Nagaraj, Bhatia, Sugandha, Monkman, James, Blick, Tony, Duijf, Pascal HG, Nagaraj, Shivashankar H, and Thompson, Erik W
- Subjects
Cell division ,RNA-sequencing ,lcsh:Medicine ,Article ,whole exome sequencing ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,proteomics ,Chromosome instability ,mesenchymal–epithelial transition (MET) ,Medicine ,Mesenchymal–epithelial transition ,epithelial–mesenchymal transition (EMT) ,030304 developmental biology ,0303 health sciences ,copy number variations (CNV) ,karyotyping ,metabolism ,seahorse extracellular flux analyser ,Cluster of differentiation ,business.industry ,lcsh:R ,Mesenchymal stem cell ,Karyotype ,General Medicine ,epithelial-mesenchymal transition (EMT) ,Chromosome 3 ,030220 oncology & carcinogenesis ,Cancer research ,business - Abstract
Epithelial−mesenchymal plasticity (EMP), encompassing epithelial−mesenchymal transition (EMT) and mesenchymal−epithelial transition (MET), are considered critical events for cancer metastasis. We investigated chromosomal heterogeneity and chromosomal instability (CIN) profiles of two sister PMC42 breast cancer (BC) cell lines to assess the relationship between their karyotypes and EMP phenotypic plasticity. Karyotyping by GTG banding and exome sequencing were aligned with SWATH quantitative proteomics and existing RNA-sequencing data from the two PMC42 cell lines; the mesenchymal, parental PMC42-ET cell line and the spontaneously epithelially shifted PMC42-LA daughter cell line. These morphologically distinct PMC42 cell lines were also compared with five other BC cell lines (MDA-MB-231, SUM-159, T47D, MCF-7 and MDA-MB-468) for their expression of EMP and cell surface markers, and stemness and metabolic profiles. The findings suggest that the epithelially shifted cell line has a significantly altered ploidy of chromosomes 3 and 13, which is reflected in their transcriptomic and proteomic expression profiles. Loss of the TGFβR2 gene from chromosome 3 in the epithelial daughter cell line inhibits its EMT induction by TGF-β stimulus. Thus, integrative ‘omics’ characterization established that the PMC42 system is a relevant MET model and provides insights into the regulation of phenotypic plasticity in breast cancer.
- Published
- 2019
44. COMMD1, from the Repair of DNA Double Strand Breaks, to a Novel Anti-Cancer Therapeutic Target
- Author
-
Pascal H.G. Duijf, Amila Suraweera, Cheng Liu, Karsten Schrobback, Mark N. Adams, Kenneth J. O'Byrne, Christian Jekimovs, Derek J. Richard, Suraweera, Amila, Duijf, Pascal HG, Jekimovs, Christian, Schrobback, Karsten, Liu, Cheng, Adams, Mark N, O'Byrne, Kenneth J, and Richard, Derek J
- Subjects
0301 basic medicine ,Cancer Research ,DNA repair ,Biology ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,medicine ,novel therapeutic target ,Viability assay ,Lung cancer ,non-small cell lung cancer ,Tissue microarray ,Cell growth ,Cancer ,COMMD1 ,Cell cycle ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,genomic stability ,medicine.disease ,respiratory tract diseases ,030104 developmental biology ,Oncology ,DNA double strand break repair ,030220 oncology & carcinogenesis ,Cancer research ,Homologous recombination - Abstract
Simple Summary Lung cancer is the most commonly diagnosed cancer worldwide and additionally the most common cause of death from cancer, with non-small cell lung cancers (NSCLC) being the most commonly diagnosed form of the disease. As drug resistance is a key issue halting chemotherapy effectiveness, there is a great need to identify new therapeutic targets. The aims of this study were to investigate the function of the protein, COMMD1, in the repair of DNA double strand breaks and the therapeutic potential of COMMD1 in NSCLC. Here, we demonstrate for the first time how an additional COMMD family member, COMMD1, functions in the repair of DNA double strand breaks and may be relevant as a therapeutic target and prognostic factor in NSCLC. These novel findings highlight the potential of a novel approach to NSCLC therapy, by targeting an overexpressed protein. Abstract Lung cancer has the highest incidence and mortality among all cancers, with non-small cell lung cancer (NSCLC) accounting for 85–90% of all lung cancers. Here we investigated the function of COMMD1 in the repair of DNA double strand breaks (DSBs) and as a prognostic and therapeutic target in NSCLC. COMMD1 function in DSB repair was investigated using reporter assays in COMMD1-siRNA-depleted cells. The role of COMMD1 in NSCLC was investigated using bioinformatic analysis, qRT-PCR and immunoblotting of control and NSCLC cells, tissue microarrays, cell viability and cell cycle experiments. DNA repair assays demonstrated that COMMD1 is required for the efficient repair of DSBs and reporter assays showed that COMMD1 functions in both non-homologous-end-joining and homologous recombination. Bioinformatic analysis showed that COMMD1 is upregulated in NSCLC, with high levels of COMMD1 associated with poor patient prognosis. COMMD1 mRNA and protein were upregulated across a panel of NSCLC cell lines and siRNA-mediated depletion of COMMD1 decreased cell proliferation and reduced cell viability of NSCLC, with enhanced death after exposure to DNA damaging-agents. Bioinformatic analyses demonstrated that COMMD1 levels positively correlate with the gene ontology DNA repair gene set enrichment signature in NSCLC. Taken together, COMMD1 functions in DSB repair, is a prognostic maker in NSCLC and is potentially a novel anti-cancer therapeutic target for NSCLC.
- Published
- 2021
- Full Text
- View/download PDF
45. CEP55 is a determinant of cell fate during perturbed mitosis in breast cancer
- Author
-
Katia Nones, Prahlad V. Raninga, Paul Timpson, Nicola Waddell, Jessie Jeffery, Sriganesh Srihari, Sunil R. Lakhani, Winnie Fernando, Pascal H.G. Duijf, Devathri Nanayakkara, J. Alejandro Lopez, Brian Gabrielli, Debottam Sinha, Jodi M. Saunus, Claire Vennin, Kevin J. Spring, Murugan Kalimutho, Deepak Mittal, Kum Kum Khanna, Kalimutho, Murugan, Sinha, Debottam, Jeffery, Jessie, Nones, Katia, Srihari, Sriganesh, Fernando, Winnie C, Duijf, Pascal HG, Vennin, Claire, Raninga, Prahlad, Nanayakkara, Devathri, Mittal, Deepak, Saunus, Jodi M, Lakhani, Sunil R, López, J Alejandro, Spring, Kevin J, Timpson, Paul, Gabrielli, Brian, Waddell, Nicola, and Khanna, Kum Kum
- Subjects
0301 basic medicine ,Genome instability ,Medicine (General) ,centrosomal protein ,Cyclin B ,Gene Expression ,Mitosis ,Breast Neoplasms ,Cell Cycle Proteins ,QH426-470 ,Biology ,Cell fate determination ,Models, Biological ,03 medical and health sciences ,R5-920 ,Breast cancer ,breast cancer ,Cell Line, Tumor ,CDC2 Protein Kinase ,Genetics ,medicine ,Humans ,aneuploidy ,Pharmacology & Drug Discovery ,CEP55 ,Research Articles ,Cytokinesis ,Cancer ,Cyclin-dependent kinase 1 ,Cell Death ,Nuclear Proteins ,medicine.disease ,Aneuploidy ,genomic instability ,3. Good health ,030104 developmental biology ,Medicine, Research & Experimental ,Caspases ,Gene Knockdown Techniques ,Cancer research ,biology.protein ,Molecular Medicine ,Research Article - Abstract
The centrosomal protein, CEP55, is a key regulator of cytokinesis, and its overexpression is linked to genomic instability, a hallmark of cancer. However, the mechanism by which it mediates genomic instability remains elusive. Here, we showed that CEP55 overexpression/knockdown impacts survival of aneuploid cells. Loss of CEP55 sensitizes breast cancer cells to anti-mitotic agents through premature CDK1/cyclin B activation and CDK1 caspase-dependent mitotic cell death. Further, we showed that CEP55 is a downstream effector of the MEK1/2-MYC axis. Blocking MEK1/2-PLK1 signaling therefore reduced outgrowth of basal-like syngeneic and human breast tumors in invi v models. In conclusion, high CEP55 levels dictate cell fate during perturbed mitosis. Forced mitotic cell death by blocking MEK1/2-PLK1 represents a potential therapeutic strategy for MYC-CEP55-dependent basal-like, triple-negative breast cancers. Refereed/Peer-reviewed
- Published
- 2018
46. Translocation Breakpoints Preferentially Occur in Euchromatin and Acrocentric Chromosomes
- Author
-
Pascal H.G. Duijf, John P. Grady, Cheng Yu Lin, Ankit Shukla, Eloise Dray, J. Lynn Fink, Lin, Cheng-Yu, Shukla, Ankit, Grady, John P, Fink, J Lynn, Dray, Eloise, and Duijf, Pascal HG
- Subjects
0301 basic medicine ,Cancer Research ,Euchromatin ,DNA repair ,cohesin ,Chromosomal translocation ,lymphoma ,Biology ,lcsh:RC254-282 ,Article ,Fusion gene ,03 medical and health sciences ,Centromere ,translocations ,DNA double strand breaks ,leukemia ,V(D)J recombination ,CTCF ,nucleolus ,acrocentric chromosomes ,Genetics ,Nucleoplasm ,Karyotype ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Chromatin ,030104 developmental biology ,Oncology - Abstract
Chromosomal translocations drive the development of many hematological and some solid cancers. Several factors have been identified to explain the non-random occurrence of translocation breakpoints in the genome. These include chromatin density, gene density and CCCTC-binding factor (CTCF)/cohesin binding site density. However, such factors are at least partially interdependent. Using 13,844 and 1563 karyotypes from human blood and solid cancers, respectively, our multiple regression analysis only identified chromatin density as the primary statistically significant predictor. Specifically, translocation breakpoints preferentially occur in open chromatin. Also, blood and solid tumors show markedly distinct translocation signatures. Strikingly, translocation breakpoints occur significantly more frequently in acrocentric chromosomes than in non-acrocentric chromosomes. Thus, translocations are probably often generated around nucleoli in the inner nucleoplasm, away from the nuclear envelope. Importantly, our findings remain true both in multivariate analyses and after removal of highly recurrent translocations. Finally, we applied pairwise probabilistic co-occurrence modeling. In addition to well-known highly prevalent translocations, such as those resulting in BCR-ABL1 (BCR-ABL) and RUNX1-RUNX1T1 (AML1-ETO) fusion genes, we identified significantly underrepresented translocations with putative fusion genes, which are probably subject to strong negative selection during tumor evolution. Taken together, our findings provide novel insights into the generation and selection of translocations during cancer development. Refereed/Peer-reviewed
- Published
- 2018
47. Contradictory mRNA and protein misexpression of EEF1A1 in ductal breast carcinoma due to cell cycle regulation and cellular stress
- Author
-
Cheng Yu Lin, Pascal H.G. Duijf, Alexandra Beattie, Behzad Baradaran, Eloise Dray, Lin, Cheng-Yu, Beattie, Alexandra, Baradaran, Behzad, Dray, Eloise, and Duijf, Pascal HG
- Subjects
0301 basic medicine ,Cell Survival ,Cell ,lcsh:Medicine ,Breast Neoplasms ,Kaplan-Meier Estimate ,Biology ,carcinoma ,03 medical and health sciences ,Peptide Elongation Factor 1 ,Carcinoma ,medicine ,Biomarkers, Tumor ,Humans ,RNA, Messenger ,Heat shock ,lcsh:Science ,Alleles ,breast ,Multidisciplinary ,Cell growth ,Carcinoma, Ductal, Breast ,Cell Cycle ,lcsh:R ,Cell cycle ,medicine.disease ,ductal ,Eukaryotic translation elongation factor 1 alpha 1 ,Ductal Breast Carcinoma ,Multidisciplinary Sciences ,Oxidative Stress ,030104 developmental biology ,medicine.anatomical_structure ,Receptors, Estrogen ,Cancer cell ,Cancer research ,Female ,lcsh:Q ,cell cycle ,EEF1A1 protein ,Heat-Shock Response - Abstract
Encoded by EEF1A1, the eukaryotic translation elongation factor eEF1α1 strongly promotes the heat shock response, which protects cancer cells from proteotoxic stress, following for instance oxidative stress, hypoxia or aneuploidy. Unexpectedly, therefore, we find that EEF1A1 mRNA levels are reduced in virtually all breast cancers, in particular in ductal carcinomas. Univariate and multivariate analyses indicate that EEF1A1 mRNA underexpression independently predicts poor patient prognosis for estrogen receptor-positive (ER+) cancers. EEF1A1 mRNA levels are lowest in the most invasive, lymph node-positive, advanced stage and postmenopausal tumors. In sharp contrast, immunohistochemistry on 100 ductal breast carcinomas revealed that at the protein level eEF1α1 is ubiquitously overexpressed, especially in ER+ , progesterone receptor-positive and lymph node-negative tumors. Explaining this paradox, we find that EEF1A1 mRNA levels in breast carcinomas are low due to EEF1A1 allelic copy number loss, found in 27% of tumors, and cell cycle-specific expression, because mRNA levels are high in G1 and low in proliferating cells. This also links estrogen-induced cell proliferation to clinical observations. In contrast, high eEF1α1 protein levels protect tumor cells from stress-induced cell death. These observations suggest that, by obviating EEF1A1 transcription, cancer cells can rapidly induce the heat shock response following proteotoxic stress, and survive.
- Published
- 2018
48. Overexpression of the E2F target gene CENPI promotes chromosome instability and predicts poor prognosis in estrogen receptor-positive breast cancer
- Author
-
Cheng Yu Lin, Pascal H.G. Duijf, Eloise Dray, Thu H.M. Nguyen, Pulari U. Thangavelu, Srividya Vaidyanathan, Thangavelu, Pulari U, Lin, Cheng-Yu, Vaidyanathan, Srividya, Nguyen, Thu HM, Dray, Eloise, and Duijf, Pascal HG
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,CENPI ,Aneuploidy ,Estrogen receptor ,Biology ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,breast cancer ,Chromosome instability ,medicine ,aneuploidy ,skin and connective tissue diseases ,Mitosis ,Cell Biology ,medicine.disease ,Spindle checkpoint ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,prognosis ,Carcinogenesis ,Breast carcinoma ,chromosome instability - Abstract
During cell division, chromosome segregation is facilitated by the mitotic checkpoint, or spindle assembly checkpoint (SAC), which ensures correct kinetochore-microtubule attachments and prevents premature sister-chromatid separation. It is well established that misexpression of SAC components on the outer kinetochores promotes chromosome instability (CIN) and tumorigenesis. Here, we study the expression of CENP-I, a key component of the HIKM complex at the inner kinetochores, in breast cancer, including ductal, lobular, medullary and male breast carcinomas. CENPI mRNA and protein levels are significantly elevated in estrogen receptor-positive (ER+) but not in estrogen receptor-negative (ER-) breast carcinoma. Well-established prognostic tests indicate that CENPI overexpression constitutes a powerful independent marker for poor patient prognosis and survival in ER+ breast cancer. We further demonstrate that CENPI is an E2F target gene. Consistently, it is overexpressed in RB1-deficient breast cancers. However, CENP-I overexpression is not purely due to cell cycle-associated expression. In ER+ breast cancer cells, CENP-I overexpression promotes CIN, especially chromosome gains. In addition, in ER+ breast carcinomas the degree of CENPI overexpression is proportional to the level of aneuploidy and CENPI overexpression is one of the strongest markers for CIN identified to date. Our results indicate that overexpression of the inner kinetochore protein CENP-I promotes CIN and forecasts poor prognosis for ER+ breast cancer patients. These observations provide novel mechanistic insights and have important implications for breast cancer diagnostics and potentially therapeutic targeting. Refereed/Peer-reviewed
- Published
- 2017
49. In epithelial cancers, aberrant COL17A1 promoter methylation predicts its misexpression and increased invasion
- Author
-
Pascal H.G. Duijf, Pulari U. Thangavelu, Tibor Krenács, Eloise Dray, Thangavelu, Pulari U, Krenács, Tibor, Dray, Eloise, and Duijf, Pascal HG
- Subjects
0301 basic medicine ,Uterine Cervical Neoplasms ,Breast Neoplasms ,Biology ,Autoantigens ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,breast cancer ,Invasion ,Genetics ,medicine ,Humans ,Neoplasm Invasiveness ,Neoplasms, Glandular and Epithelial ,Promoter Regions, Genetic ,Molecular Biology ,Genetics (clinical) ,Collagen XVII ,collagen XVII ,Genetics & Heredity ,epigenetics ,Research ,Intravasation ,Cancer ,DNA Methylation ,Non-Fibrillar Collagens ,medicine.disease ,invasion ,Prognosis ,Head and neck squamous-cell carcinoma ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,DNA methylation ,Cancer research ,Cervical cancer ,Adenocarcinoma ,Epigenetics ,Female ,prognosis ,Developmental Biology - Abstract
Background Metastasis is a leading cause of death among cancer patients. In the tumor microenvironment, altered levels of extracellular matrix proteins, such as collagens, can facilitate the first steps of cancer cell metastasis, including invasion into surrounding tissue and intravasation into the blood stream. However, the degree of misexpression of collagen genes in tumors remains understudied, even though this knowledge could greatly facilitate the development of cancer treatment options aimed at preventing metastasis. Methods We systematically evaluate the expression of all 44 collagen genes in breast cancer and assess whether their misexpression provides clinical prognostic significance. We use immunohistochemistry on 150 ductal breast cancers and 361 cervical cancers and study DNA methylation in various epithelial cancers. Results In breast cancer, various tests show that COL4A1 and COL4A2 overexpression and COL17A1 (BP180, BPAG2) underexpression provide independent prognostic strength (HR = 1.25, 95% CI = 1.17–1.34, p = 3.03 × 10−10; HR = 1.18, 95% CI = 1.11–1.25, p = 8.11 × 10−10; HR = 0.86, 95% CI = 0.81–0.92, p = 4.57 × 10−6; respectively). Immunohistochemistry on ductal breast cancers confirmed that the COL17A1 protein product, collagen XVII, is underexpressed. This strongly correlates with advanced stage, increased invasion, and postmenopausal status. In contrast, immunohistochemistry on cervical tumors showed that collagen XVII is overexpressed in cervical cancer and this is associated with increased local dissemination. Interestingly, consistent with the opposed direction of misexpression in these cancers, the COL17A1 promoter is hypermethylated in breast cancer and hypomethylated in cervical cancer. We also find that the COL17A1 promoter is hypomethylated in head and neck squamous cell carcinoma, lung squamous cell carcinoma, and lung adenocarcinoma, in all of which collagen XVII overexpression has previously been shown. Conclusions Paradoxically, collagen XVII is underexpressed in breast cancer and overexpressed in cervical and other epithelial cancers. However, the COL17A1 promoter methylation status accurately predicts both the direction of misexpression and the increased invasive nature for five out of five epithelial cancers. This implies that aberrant epigenetic control is a key driver of COL17A1 gene misexpression and tumor cell invasion. These findings have significant clinical implications, suggesting that the COL17A1 promoter methylation status can be used to predict patient outcome. Moreover, epigenetic targeting of COL17A1 could represent a novel strategy to prevent metastasis in patients. Electronic supplementary material The online version of this article (doi:10.1186/s13148-016-0290-6) contains supplementary material, which is available to authorized users.
- Published
- 2016
50. Overexpression of Ran GTPase Components Regulating Nuclear Export, but not Mitotic Spindle Assembly, Marks Chromosome Instability and Poor Prognosis in Breast Cancer
- Author
-
Pulari U. Thangavelu, Srividya Vaidyanathan, Pascal H.G. Duijf, Vaidyanathan, Srividya, Thangavelu, Pulari U, and Duijf, Pascal HG
- Subjects
0301 basic medicine ,Cancer Research ,metasasis ,Mitosis ,Breast Neoplasms ,Spindle Apparatus ,XPO5 ,gene-expression profiles ,GTP Phosphohydrolases ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Chromosomal Instability ,medicine ,Humans ,Pharmacology (medical) ,Nuclear protein ,Nuclear export signal ,molecular subtypes ,business.industry ,Nuclear Proteins ,adjuvant therapy ,medicine.disease ,Prognosis ,Survival Analysis ,Spindle apparatus ,Cell biology ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Ran ,Cancer research ,Female ,Nuclear transport ,business - Abstract
Background: Ran GTPase regulates nuclear import, nuclear export, and mitotic spindle assembly. The multifunctional involvement of seventeen Ran GTPase components in these processes has complicated research into how each contributes to cancer development. Objective: To assess whether individual and process-specific misexpression of Ran GTPase components contribute to chromosome instability (CIN) and worsen breast cancer patient prognosis. Methods: Using publicly available datasets, we studied the degree of misexpression of all Ran GTPase signaling components in breast cancer, assessed their involvement in CIN and used four clinical tests to evaluate whether their misregulation may constitute independent prognostic predictors. Results: A significant majority of Ran GTPase signaling components is overexpressed in breast cancer. Strikingly, spindle assembly components are overexpressed and associated with CIN with only marginal significance and four independent tests indicate that this does not worsen patient outcome. Overexpression of nuclear import components is neither CIN-associated nor clinically significant. In sharp contrast, overexpression of nuclear export components constitutes a strong independent marker for both CIN and poor patient prognosis. We identify Exportin 2/CSE1L, Exportin 3/XPOT, Exportin 5/XPO5, and RANBP1 as novel potential targets. Conclusions: We find that overexpression of Ran GTPase components involved in nuclear export, but not nuclear import or mitotic spindle assembly, is a strong CIN-associated marker for poor breast cancer prognosis. This could mean that increased nuclear export (of, for instance, pRb, p53, p73, BRCA1, p21, p27, E2F4, IκB, survivin), rather than spindle defects, mainly drives CIN and tumorigenesis. Hence, selective inhibitors of nuclear export may be effective for treating the most aggressive and chromosomally unstable breast cancers. Refereed/Peer-reviewed
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.