Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Cheryl A. Dean is active.

Publication


Featured researches published by Cheryl A. Dean.


Stem Cells | 2011

Aldehyde Dehydrogenase Activity of Breast Cancer Stem Cells Is Primarily Due To Isoform ALDH1A3 and Its Expression Is Predictive of Metastasis

Paola Marcato; Cheryl A. Dean; Da Pan; Rakhna Araslanova; Megan Gillis; Madalsa Joshi; Lucy Helyer; Lu-Zhe Pan; Andrew M. Leidal; Shashi Gujar; Carman A. Giacomantonio; Patrick W.K. Lee

Cancer stem cells (CSCs) are proposed to initiate cancer and propagate metastasis. Breast CSCs identified by aldehyde dehydrogenase (ALDH) activity are highly tumorigenic in xenograft models. However, in patient breast tumor immunohistological studies, where CSCs are identified by expression of ALDH isoform ALDH1A1, CSC prevalence is not correlative with metastasis, raising some doubt as to the role of CSCs in cancer. We characterized the expression of all 19 ALDH isoforms in patient breast tumor CSCs and breast cancer cell lines by total genome microarray expression analysis, immunofluorescence protein expression studies, and quantitative polymerase chain reaction. These studies revealed that ALDH activity of patient breast tumor CSCs and cell lines correlates best with expression of another isoform, ALDH1A3, not ALDH1A1. We performed shRNA knockdown experiments of the various ALDH isoforms and found that only ALDH1A3 knockdown uniformly reduced ALDH activity of breast cancer cells. Immunohistological studies with fixed patient breast tumor samples revealed that ALDH1A3 expression in patient breast tumors correlates significantly with tumor grade, metastasis, and cancer stage. Our results, therefore, identify ALDH1A3 as a novel CSC marker with potential clinical prognostic applicability, and demonstrate a clear correlation between CSC prevalence and the development of metastatic breast cancer. STEM CELLS 2011;29:32–45


Cell Cycle | 2011

Aldehyde dehydrogenase: Its role as a cancer stem cell marker comes down to the specific isoform

Paola Marcato; Cheryl A. Dean; Carman A. Giacomantonio; Patrick W.K. Lee

Recent evidence suggests that enhanced aldehyde dehydrogenase (ALDH) activity is a hallmark of cancer stem cells (CSC) measurable by the aldefluor assay. ALDH1A1, one of 19 ALDH isoforms expressed in humans, was generally believed to be responsible for the ALDH activity of CSCs. More recently, experiments with murine hematopoietic stem cells, murine progenitor pancreatic cells, and human breast CSCs indicate that other ALDH isoforms, particularly ALDH1A3, significantly contribute to aldefluor positivity, which may be tissue and cancer specific. Therefore, potential prognostic application involving the use of CSC prevalence in tumor tissue to predict patient outcome requires the identification and quantification of specific ALDH isoforms. Herein we review the suggested roles of ALDH in CSC biology and the immunohistological studies testing the potential application of ALDH isoforms as novel cancer prognostic indicators.


Molecular Therapy | 2009

Oncolytic Reovirus Effectively Targets Breast Cancer Stem Cells

Paola Marcato; Cheryl A. Dean; Carman A. Giacomantonio; Patrick W.K. Lee

Recent evidence suggests that cancer stem cells (CSCs) play an important role in cancer, as these cells possess enhanced tumor-forming capabilities and are resistant to current anticancer therapies. Hence, novel cancer therapies will need to be tested for both tumor regression and CSC targeting. Herein we show that oncolytic reovirus that induces regression of human breast cancer primary tumor samples xenografted in immunocompromised mice also effectively targets and kills CSCs in these tumors. CSCs were identified based on CD24(-)CD44(+) cell surface expression and overexpression of aldehyde dehydrogenase. Upon reovirus treatment, the CSC population was reduced at the same rate as non-CSCs within the tumor. Immunofluorescence of breast tumor tissue samples from the reovirus- and mock-treated mice confirmed that both CSCs and non-CSCs were infectible by reovirus, and terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling (TUNEL) assay showed that both populations died by apoptosis. Ras, which has been shown to mediate reovirus oncolysis, was found to be present at similar levels in all cell types, and this is consistent with their comparable sensitivity to reovirus. These experiments indicate that oncolytic reovirus has the potential to induce tumor regression in breast cancer patients. More important, the CSC population was equally reduced and was as susceptible to reovirus treatment as the non-CSC population.


Neoplasia | 2014

Core needle biopsy of breast cancer tumors increases distant metastases in a mouse model.

Edward G. Mathenge; Cheryl A. Dean; Derek Clements; Steffany Photopoulos; Krysta Mila Coyle; Michael Giacomantonio; Benjamin Malueth; Anna Nunokawa; J.L. Jordan; John D. Lewis; Shashi Gujar; Paola Marcato; Patrick W.K. Lee; Carman A. Giacomantonio

INTRODUCTION: Incisional biopsies, including the diagnostic core needle biopsy (CNB), routinely performed before surgical excision of breast cancer tumors are hypothesized to increase the risk of metastatic disease. In this study, we experimentally determined whether CNB of breast cancer tumors results in increased distant metastases and examine important resultant changes in the primary tumor and tumor microenvironment associated with this outcome. METHOD: To evaluate the effect of CNB on metastasis development, we implanted murine mammary 4T1 tumor cells in BALB/c mice and performed CNB on palpable tumors in half the mice. Subsequently, emulating the human scenario, all mice underwent complete tumor excision and were allowed to recover, with attendant metastasis development. Tumor growth, lung metastasis, circulating tumor cell (CTC) levels, variation in gene expression, composition of the tumor microenvironment, and changes in immunologic markers were compared in biopsied and non-biopsied mice. RESULTS: Mice with biopsied tumors developed significantly more lung metastases compared to non-biopsied mice. Tumors from biopsied mice contained a higher frequency of myeloid-derived suppressor cells (MDSCs) accompanied by reduced CD4 + T cells, CD8 + T cells, and macrophages, suggesting biopsy-mediated development of an increasingly immunosuppressive tumor microenvironment. We also observed a CNB-dependent up-regulation in the expression of SOX4, Ezh2, and other key epithelial-mesenchymal transition (EMT) genes, as well as increased CTC levels among the biopsy group. CONCLUSION: CNB creates an immunosuppressive tumor microenvironment, increases EMT, and facilitates release of CTCs, all of which likely contribute to the observed increase in development of distant metastases.


Cell Death & Differentiation | 2016

The NAD + salvage pathway modulates cancer cell viability via p73

Tanveer Sharif; D-G Ahn; R-Z Liu; E Pringle; Emma Martell; Cathleen Dai; Anna Nunokawa; M Kwak; Derek Clements; J P Murphy; Cheryl A. Dean; Paola Marcato; C McCormick; Roseline Godbout; Shashi Gujar; Patrick W.K. Lee

The involvement of the nicotinamide adenine dinucleotide (NAD+) salvage pathway in cancer cell survival is poorly understood. Here we show that the NAD+ salvage pathway modulates cancer cell survival through the rarely mutated tumour suppressor p73. Our data show that pharmacological inhibition or knockdown of nicotinamide phosphoribosyltransferase (NAMPT), a rate-limiting enzyme in the NAD+ salvage pathway, enhances autophagy and decreases survival of cancer cells in a p53-independent manner. Such NAMPT inhibition stabilizes p73 independently of p53 through increased acetylation and decreased ubiquitination, resulting in enhanced autophagy and cell death. These effects of NAMPT inhibition can be effectively reversed using nicotinamide mononucleotide (NMN), the enzymatic product of NAMPT. Similarly, knockdown of p73 also decreases NAMPT inhibition-induced autophagy and cell death, whereas overexpression of p73 alone enhances these effects. We show that the breast cancer cell lines (MCF-7, MDA-MB-231 and MDA-MB-468) harbour significantly higher levels of NAMPT and lower levels of p73 than does the normal cell line (MCF-10A), and that NAMPT inhibition is cytotoxic exclusively to the cancer cells. Furthermore, data from 176 breast cancer patients demonstrate that higher levels of NAMPT and lower levels of p73 correlate with poorer patient survival, and that high-grade tumours have significantly higher NAMPT/p73 mRNA ratios. Therefore, the inverse relationship between NAMPT and p73 demonstrable in vitro is also reflected from the clinical data. Taken together, our studies reveal a new NAMPT-p73 nexus that likely has important implications for cancer diagnosis, prognosis and treatment.


Journal of Immunology | 2015

Newly Recruited CD11b+, GR-1+, Ly6Chigh Myeloid Cells Augment Tumor-Associated Immunosuppression Immediately following the Therapeutic Administration of Oncolytic Reovirus

Derek Clements; Andra Sterea; Youra Kim; Erin Helson; Cheryl A. Dean; Anna Nunokawa; Krysta Mila Coyle; Tanveer Sharif; Paola Marcato; Shashi Gujar; Patrick W.K. Lee

Tumor-associated immunosuppression aids cancer cells to escape immune-mediated attack and subsequent elimination. Recently, however, many oncolytic viruses, including reovirus, have been reported to overturn such immunosuppression and promote the development of a clinically desired antitumor immunity, which is known to promote favorable patient outcomes. Contrary to this existing paradigm, in this article we demonstrate that reovirus augments tumor-associated immunosuppression immediately following its therapeutic administration. Our data show that reovirus induces preferential differentiation of highly suppressive CD11b+, Gr-1+, Ly6Chigh myeloid cells from bone marrow hematopoietic progenitor cells. Furthermore, reovirus administration in tumor-bearing hosts drives time-dependent recruitment of CD11b+, Gr-1+, Ly6Chigh myeloid cells in the tumor milieu, which is further supported by virus-induced increased expression of numerous immune factors involved in myeloid-derived suppressor cell survival and trafficking. Most importantly, CD11b+, Gr-1+, Ly6Chigh myeloid cells specifically potentiate the suppression of T cell proliferation and are associated with the absence of IFN-γ response in the tumor microenvironment early during oncotherapy. Considering that the qualitative traits of a specific antitumor immunity are largely dictated by the immunological events that precede its development, our findings are of critical importance and must be considered while devising complementary interventions aimed at promoting the optimum efficacy of oncolytic virus–based anticancer immunotherapies.


Oncotarget | 2016

Breast cancer subtype dictates DNA methylation and ALDH1A3-mediated expression of tumor suppressor RARRES1

Krysta Mila Coyle; J. Patrick Murphy; Dejan Vidovic; Cheryl A. Dean; Mohammad Sultan; Derek Clements; Melissa Wallace; Margaret L. Thomas; Amos S. Hundert; Carman A. Giacomantonio; Lucy Helyer; Shashi Gujar; Patrick W.K. Lee; Ian C. G. Weaver; Paola Marcato

Breast cancer subtyping, based on the expression of hormone receptors and other genes, can determine patient prognosis and potential options for targeted therapy. Among breast cancer subtypes, tumors of basal-like and claudin-low subtypes are typically associated with worse patient outcomes, are primarily classified as triple-negative breast cancers (TNBC), and cannot be treated with existing hormone-receptor-targeted therapies. Understanding the molecular basis of these subtypes will lead to the development of more effective treatment options for TNBC. In this study, we focus on retinoic acid receptor responder 1 (RARRES1) as a paradigm to determine if breast cancer subtype dictates protein function and gene expression regulation. Patient tumor dataset analysis and gene expression studies of a 26 cell-line panel, representing the five breast cancer subtypes, demonstrate that RARRES1 expression is greatest in basal-like TNBCs. Cell proliferation and tumor growth assays reveal that RARRES1 is a tumor suppressor in TNBC. Furthermore, gene expression studies, Illumina HumanMethylation450 arrays, and chromatin immunoprecipitation demonstrate that expression of RARRES1 is retained in basal-like breast cancers due to hypomethylation of the promoter. Additionally, expression of the cancer stem cell marker, aldehyde dehydrogenase 1A3, which provides the required ligand (retinoic acid) for RARRES1 transcription, is also specific to the basal-like subtype. We functionally demonstrate that the combination of promoter methylation and retinoic acid signaling dictates expression of tumor suppressor RARRES1 in a subtype-specific manner. These findings provide a precedent for a therapeutically-inducible tumor suppressor and suggest novel avenues of therapeutic intervention for patients with basal-like breast cancer.


Stem Cells | 2018

Epigenetic Silencing of TAP1 in Aldefluor+ Breast Cancer Stem Cells Contributes to Their Enhanced Immune Evasion

Mohammad Sultan; Dejan Vidovic; Arianne S. Paine; Thomas Tan Huynh; Krysta Mila Coyle; Margaret L. Thomas; Brianne Cruickshank; Cheryl A. Dean; Derek Clements; Youra Kim; Kristen Lee; Shashi Gujar; Ian C. G. Weaver; Paola Marcato

Avoiding detection and destruction by immune cells is key for tumor initiation and progression. The important role of cancer stem cells (CSCs) in tumor initiation has been well established, yet their ability to evade immune detection and targeting is only partly understood. To investigate the ability of breast CSCs to evade immune detection, we identified a highly tumorigenic population in a spontaneous murine mammary tumor based on increased aldehyde dehydrogenase activity. We performed tumor growth studies in immunocompetent and immunocompromised mice. In immunocompetent mice, growth of the spontaneous mammary tumor was restricted; however, the Aldefluor+ population was expanded, suggesting inherent resistance mechanisms. Gene expression analysis of the sorted tumor cells revealed that the Aldefluor+ tumor cells has decreased expression of transporter associated with antigen processing (TAP) genes and co‐stimulatory molecule CD80, which would decrease susceptibility to T cells. Similarly, the Aldefluor+ population of patient tumors and 4T1 murine mammary cells had decreased expression of TAP and co‐stimulatory molecule genes. In contrast, breast CSCs identified by CD44+CD24− do not have decreased expression of these genes, but do have increased expression of C‐X‐C chemokine receptor type 4. Decitabine treatment and bisulfite pyrosequencing suggests that DNA hypermethylation contributes to decreased TAP gene expression in Aldefluor+ CSCs. TAP1 knockdown resulted in increased tumor growth of 4T1 cells in immunocompetent mice. Together, this suggests immune evasion mechanisms in breast CSCs are marker specific and epigenetic silencing of TAP1 in Aldefluor+ breast CSCs contributes to their enhanced survival under immune pressure. Stem Cells 2018;36:641–654


Epigenetic Mechanisms | 2018

PO-372 Investigating the epigenetic changes underlying combination treatment of acute promyelocytic leukaemia with all-trans retinoic acid and arsenic trioxide

Thomas Tan Huynh; Dejan Vidovic; Cheryl A. Dean; Kristen Lee; Ian C. G. Weaver; Paola Marcato

Introduction Acute promyelocytic leukaemia (APL) results in the interrupted differentiation of granulocytes. Combination induction therapy using all-trans retinoic acid (ATRA) and arsenic trioxide (ATO) is the most effective treatment of APL. DNA methylation and histone modifications are epigenetic mechanisms of gene regulation aberrant in APL. While ATRA treatment of APL NB4 cells restores granulocyte differentiation and induces significant genome-wide changes in histone modifications, it does not affect the aberrant DNA methylation. Similarly, ATO has comparable effects. Therefore, we hypothesise that the increased effectiveness of ATRA and ATO when combined is due to epigenetic changes at both the histone and DNA methylation level, resulting in profound changes in gene expression. Material and methods We identified the concentration of ATRA and ATO in combination that induced enhanced differentiation and cell death of NB4 cells in comparison to single agent treatment through fluorescence activated cells sorting (FACS) of CD11b and 7-AAD labelled cells at 72 hours and 96 hours post treatment termination. Quantitative PCR (qPCR) was performed to determine the expression levels of a panel of genes after 72 hours of treatment and 96 hours post treatment termination. Results and discussions ATRA and ATO in combination induced significant differentiation and cell death of NB4 cells after 72 hours. Interestingly, only the combination-treated cells maintained differentiation when treatment was terminated for 96 hours. Additionally,combination treatment significantly increased the expression of several genes important in APL compared to single agent treatment after 72 hours as well as 96 hours post treatment termination. Chromatin immunoprecipitation (ChIP) is currently being utilised to investigate the enrichment of chromatin with acetylated and methylated histones, and qPCR was performed to assess the enrichment of several genes of interest. Finally, bisulfite pyrosequencing is being performed to characterise the specific methylation pattern of these genes of interest. Conclusion ATRA and ATO in combination synergistically induces the differentiation of APL NB4 cells. Together, the ChIP and bisulfite pyrosequencing experiments will enable us to characterise the effects of combination treatment at the epigenetic level. Ultimately, understanding the epigenetic changes induced by ATO and ATRA synergy can help improve treatment in the clinic.


Cancers | 2018

DNA Methylation Predicts the Response of Triple-Negative Breast Cancers to All-Trans Retinoic Acid

Krysta Mila Coyle; Cheryl A. Dean; Margaret L. Thomas; Dejan Vidovic; Carman A. Giacomantonio; Lucy Helyer; Paola Marcato

All-trans retinoic acid (atRA) regulates gene expression and is used to treat acute promyelocytic leukemia. Attempts to use atRA in breast cancer without a stratification strategy have resulted in limited overall effectiveness. To identify biomarkers for the treatment of triple-negative breast cancer (TNBC) with atRA, we characterized the effects of atRA on the tumor growth of 13 TNBC cell lines. This resulted in a range of effects that was not predictable based on previously hypothesized predictors of response, such as the levels of atRA nuclear shuttling proteins fatty acid binding protein 5 (FABP5) and cellular retinoic acid binding protein 2 (CRABP2). Transcriptional profiling revealed that atRA induced distinct gene expression changes in the sensitive versus resistant cell lines that were mostly independent of the presence of retinoic acid response elements (RAREs) or peroxisome proliferator response elements (PPREs). Given the importance of DNA methylation in regulating gene expression, we hypothesized that differential DNA methylation could predict the response of TNBCs to atRA. We identified over 1400 sites that were differentially methylated between atRA resistant and sensitive cell lines. These CpG sites predicted the response of four TNBC patient-derived xenografts to atRA, and we utilized these xenografts to refine the profile and identified that as many as 17% of TNBC patients could benefit from atRA treatment. These data illustrate that differential methylation of specific CpGs may be useful biomarkers for predicting the response of patient tumors to atRA treatment.

Collaboration


Dive into the Cheryl A. Dean's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge