Network


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

Hotspot


Dive into the research topics where Desiree H. Floyd is active.

Publication


Featured researches published by Desiree H. Floyd.


The Journal of Neuroscience | 2009

The neuronal microRNA miR-326 acts in a feedback loop with Notch and has therapeutic potential against brain tumors

Benjamin Kefas; Laurey Comeau; Desiree H. Floyd; Oleksandr Seleverstov; Jakub Godlewski; Tom Schmittgen; Jinmai Jiang; Charles G. diPierro; Yunqing Li; E. Antonio Chiocca; Jeongwu Lee; Howard A. Fine; Roger Abounader; Sean E. Lawler; Benjamin Purow

Little is known of microRNA interactions with cellular pathways. Few reports have associated microRNAs with the Notch pathway, which plays key roles in nervous system development and in brain tumors. We previously implicated the Notch pathway in gliomas, the most common and aggressive brain tumors. While investigating Notch mediators, we noted microRNA-326 was upregulated following Notch-1 knockdown. This neuronally expressed microRNA was not only suppressed by Notch but also inhibited Notch proteins and activity, indicating a feedback loop. MicroRNA-326 was downregulated in gliomas via decreased expression of its host gene. Transfection of microRNA-326 into both established and stem cell-like glioma lines was cytotoxic, and rescue was obtained with Notch restoration. Furthermore, miR-326 transfection reduced glioma cell tumorigenicity in vivo. Additionally, we found microRNA-326 partially mediated the toxic effects of Notch knockdown. This work demonstrates a microRNA-326/Notch axis, shedding light on the biology of Notch and suggesting microRNA-326 delivery as a therapy.


Cancer Discovery | 2013

Diacylglycerol kinase alpha is a critical signaling node and novel therapeutic target in glioblastoma and other cancers

Charli Dominguez; Desiree H. Floyd; Aizhen Xiao; Garrett R. Mullins; Benjamin Kefas; Wenjun Xin; Melissa N. Yacur; Roger Abounader; Jae K. Lee; Gabriela Mustata Wilson; Thurl E. Harris; Benjamin Purow

Although diacylglycerol kinase α (DGKα) has been linked to several signaling pathways related to cancer cell biology, it has been neglected as a target for cancer therapy. The attenuation of DGKα activity via DGKα-targeting siRNA and small-molecule inhibitors R59022 and R59949 induced caspase-mediated apoptosis in glioblastoma cells and in other cancers, but lacked toxicity in noncancerous cells. We determined that mTOR and hypoxia-inducible factor-1α (HIF-1α) are key targets of DGKα inhibition, in addition to its regulation of other oncogenes. DGKα regulates mTOR transcription via a unique pathway involving cyclic AMP. Finally, we showed the efficacy of DGKα inhibition with short hairpin RNA or a small-molecule agent in glioblastoma and melanoma xenograft treatment models, with growth delay and decreased vascularity. This study establishes DGKα as a central signaling hub and a promising therapeutic target in the treatment of cancer.


PLOS ONE | 2014

Novel Anti-Apoptotic MicroRNAs 582-5p and 363 Promote Human Glioblastoma Stem Cell Survival via Direct Inhibition of Caspase 3, Caspase 9, and Bim

Desiree H. Floyd; Ying Zhang; Bijan K. Dey; Benjamin Kefas; Hannah Breit; Kaitlyn Marks; Anindya Dutta; Christel Herold-Mende; Michael Synowitz; Rainer Glass; Roger Abounader; Benjamin Purow

Glioblastoma is the most common and lethal primary brain tumor. Tumor initiation and recurrence are likely caused by a sub-population of glioblastoma stem cells, which may derive from mutated neural stem and precursor cells. Since CD133 is a stem cell marker for both normal brain and glioblastoma, and to better understand glioblastoma formation and recurrence, we looked for dys-regulated microRNAs in human CD133+ glioblastoma stem cells as opposed to CD133+ neural stem cells isolated from normal human brain. Using FACS sorting of low-passage cell samples followed by microRNA microarray analysis, we found 43 microRNAs that were dys-regulated in common in three separate CD133+ human glioblastomas compared to CD133+ normal neural stem cells. Among these were several microRNAs not previously associated with cancer. We then verified the microRNAs dys-regulated in glioblastoma using quantitative real time PCR and Taqman analysis of the original samples, as well as human GBM stem cell and established cell lines and many human specimens. We show that two candidate oncogenic microRNAs, miR-363 and miR-582-5p, can positively influence glioblastoma survival, as shown by forced expression of the microRNAs and their inhibitors followed by cell number assay, Caspase 3/7 assay, Annexin V apoptosis/fluorescence activated cell sorting, siRNA rescue of microRNA inhibitor treatment, as well as 3′UTR mutagenesis to show luciferase reporter rescue of the most successful targets. miR-582-5p and miR-363 are shown to directly target Caspase 3, Caspase 9, and Bim.


Neuro-oncology | 2014

Micro-masters of glioblastoma biology and therapy: increasingly recognized roles for microRNAs

Desiree H. Floyd; Benjamin Purow

MicroRNAs are small noncoding RNAs encoded in eukaryotic genomes that have been found to play critical roles in most biological processes, including cancer. This is true for glioblastoma, the most common and lethal primary brain tumor, for which microRNAs have been shown to strongly influence cell viability, stem cell characteristics, invasiveness, angiogenesis, metabolism, and immune evasion. Developing microRNAs as prognostic markers or as therapeutic agents is showing increasing promise and has potential to reach the clinic in the next several years. This succinct review summarizes current progress and future directions in this exciting and steadily expanding field.


Integrative Biology | 2016

Interstitial flow differentially increases patient-derived glioblastoma stem cell invasion via CXCR4, CXCL12, and CD44-mediated mechanisms

Kathryn M. Kingsmore; Daniel K. Logsdon; Desiree H. Floyd; Shayn M. Peirce; Benjamin Purow; Jennifer M. Munson

Glioblastoma (GBM) prognosis remains dismal due in part to the invasiveness of GBM cells. Interstitial fluid flow (IFF) has been shown to increase invasion of glioma cells in vitro through the CXCR4 receptor interacting with autologous, pericellular gradients of CXCL12 (autologous chemotaxis) or through the CD44 receptor interactions with the extracellular matrix (hyaluronan-mediated mechanotransduction). These mechanisms have not been examined together and thus we hypothesized that both mechanisms contribute to invasion in populations of cancer cells. Therefore, we examined IFF-stimulated CXCR4-, CXCL12-, and CD44-dependent invasion in patient-derived glioblastoma stem cells (GSCs). Using our 3D in vitro assay and correlative in vivo studies we demonstrated GSC lines show increased invasion with flow. This flow-stimulated invasion was reduced by blockade of CXCR4, CXCL12, and/or CD44, revealing that GSC invasion may be mediated simultaneously by both mechanisms. Characterization of CXCR4+, CXCL12+, and CD44+ populations in four GSC lines revealed different percentages of protein positive subpopulations for each line. We developed an agent-based model to identify the contributions of each subpopulation to flow-stimulated invasion and validated the model through comparisons with experimental blocking studies. Clinically relevant radiation therapy increased flow-stimulated invasion in one GSC line. Our agent-based model predicted that IFF-stimulated invasion is driven primarily by CXCR4+CXCL12+ populations, and, indeed our irradiated cells had an increase in this subpopulation. Together, these data indicate that different mechanisms govern the flow response across GSCs, but that within a single patient, there are subpopulations of GSCs that respond to flow via either CD44- or CXCR4-CXCL12 mechanisms.


PLOS ONE | 2014

Sustained radiosensitization of hypoxic glioma cells after oxygen pretreatment in an animal model of glioblastoma and in vitro models of tumor hypoxia.

Ryon Clarke; Shayan Moosa; Matthew J. Anzivino; Yi Wang; Desiree H. Floyd; Benjamin Purow; Kevin S. Lee

Glioblastoma multiforme (GBM) is the most common and lethal form of brain cancer and these tumors are highly resistant to chemo- and radiotherapy. Radioresistance is thought to result from a paucity of molecular oxygen in hypoxic tumor regions, resulting in reduced DNA damage and enhanced cellular defense mechanisms. Efforts to counteract tumor hypoxia during radiotherapy are limited by an attendant increase in the sensitivity of healthy brain tissue to radiation. However, the presence of heightened levels of molecular oxygen during radiotherapy, while conventionally deemed critical for adjuvant oxygen therapy to sensitize hypoxic tumor tissue, might not actually be necessary. We evaluated the concept that pre-treating tumor tissue by transiently elevating tissue oxygenation prior to radiation exposure could increase the efficacy of radiotherapy, even when radiotherapy is administered after the return of tumor tissue oxygen to hypoxic baseline levels. Using nude mice bearing intracranial U87-luciferase xenografts, and in vitro models of tumor hypoxia, the efficacy of oxygen pretreatment for producing radiosensitization was tested. Oxygen-induced radiosensitization of tumor tissue was observed in GBM xenografts, as seen by suppression of tumor growth and increased survival. Additionally, rodent and human glioma cells, and human glioma stem cells, exhibited prolonged enhanced vulnerability to radiation after oxygen pretreatment in vitro, even when radiation was delivered under hypoxic conditions. Over-expression of HIF-1α reduced this radiosensitization, indicating that this effect is mediated, in part, via a change in HIF-1-dependent mechanisms. Importantly, an identical duration of transient hyperoxic exposure does not sensitize normal human astrocytes to radiation in vitro. Taken together, these results indicate that briefly pre-treating tumors with elevated levels of oxygen prior to radiotherapy may represent a means for selectively targeting radiation-resistant hypoxic cancer cells, and could serve as a safe and effective adjuvant to radiation therapy for patients with GBM.


Neuro-oncology | 2018

Targeting the mesenchymal subtype in glioblastoma and other cancers via inhibition of diacylglycerol kinase alpha

Inan Olmez; Shawn Love; Aizhen Xiao; Laryssa Manigat; Peyton Randolph; Brian D. McKenna; Brian Neal; Salome Boroda; Ming Li; Breanna Brenneman; Roger Abounader; Desiree H. Floyd; Jeongwu Lee; Ichiro Nakano; Jakub Godlewski; Agnieszka Bronisz; Erik P. Sulman; Marty W. Mayo; Daniel Gioeli; Michael J. Weber; Thurl E. Harris; Benjamin Purow

Background The mesenchymal phenotype in glioblastoma (GBM) and other cancers drives aggressiveness and treatment resistance, leading to therapeutic failure and recurrence of disease. Currently, there is no successful treatment option available against the mesenchymal phenotype. Methods We classified patient-derived GBM stem cell lines into 3 subtypes: proneural, mesenchymal, and other/classical. Each subtypes response to the inhibition of diacylglycerol kinase alpha (DGKα) was compared both in vitro and in vivo. RhoA activation, liposome binding, immunoblot, and kinase assays were utilized to elucidate the novel link between DGKα and geranylgeranyltransferase I (GGTase I). Results Here we show that inhibition of DGKα with a small-molecule inhibitor, ritanserin, or RNA interference preferentially targets the mesenchymal subtype of GBM. We show that the mesenchymal phenotype creates the sensitivity to DGKα inhibition; shifting GBM cells from the proneural to the mesenchymal subtype increases ritanserin activity, with similar effects in epithelial-mesenchymal transition models of lung and pancreatic carcinoma. This enhanced sensitivity of mesenchymal cancer cells to ritanserin is through inhibition of GGTase I and downstream mediators previously associated with the mesenchymal cancer phenotype, including RhoA and nuclear factor-kappaB. DGKα inhibition is synergistic with both radiation and imatinib, a drug preferentially affecting proneural GBM. Conclusions Our findings demonstrate that a DGKα-GGTase I pathway can be targeted to combat the treatment-resistant mesenchymal cancer phenotype. Combining therapies with greater activity against each GBM subtype may represent a viable therapeutic option against GBM.


Oncotarget | 2017

CDK4/6 inhibition is more active against the glioblastoma proneural subtype

Ming D. Li; Aizhen Xiao; Desiree H. Floyd; Inan Olmez; Jeongwu Lee; Jakub Godlewski; Agnieszka Bronisz; Krishna P.L. Bhat; Erik P. Sulman; Ichiro Nakano; Benjamin Purow

Glioblastoma (GBM) is the most common and lethal brain tumor. Gene expression profiling has classified GBM into distinct subtypes, including proneural, mesenchymal, and classical, and identifying therapeutic vulnerabilities of these subtypes is an extremely high priority. We leveraged The Cancer Genome Atlas (TCGA) data, in particular for microRNA expression, to seek druggable core pathways in GBM. The E2F1-regulated miR-17˜92 cluster and its analogs are shown to be highly expressed in proneural GBM and in GSC lines, suggesting the E2F cell cycle pathway might be a key driver in proneural GBM. Consistently, CDK4/6 inhibition with palbociclib preferentially inhibited cell proliferation in vitro in a majority of proneural GSCs versus those of other subtypes. Palbociclib treatment significantly prolonged survival of mice with established intracranial xenografts of a proneural GSC line. We show that most of these sensitive PN GSCs expressed higher levels of CDK6 and had intact Rb1, while two GSC lines with CDK4 overexpression and null Rb1 were highly resistant to palbociclib. Importantly, palbociclib treatment of proneural GSCs upregulated mesenchymal-associated markers and downregulated proneural-associated markers, suggesting that CDK4/6 inhibition induced proneural-mesenchymal transition and underscoring the enhanced role of the E2F cell cycle pathway in the proneural subtype. Lastly, the combination of palbociclib and N,N-diethylaminobenzaldehyde, an inhibitor of the mesenchymal driver ALDH1A3, showed strong synergistic inhibitory effects against proneural GSC proliferation. Taken together, our results reveal that proneural GBM has increased vulnerability to CDK4/6 inhibition, and the proneural subtype undergoes dynamic reprogramming upon palbociclib treatment—suggesting the need for a combination therapeutic strategy.Glioblastoma (GBM) is the most common and lethal brain tumor. Gene expression profiling has classified GBM into distinct subtypes, including proneural, mesenchymal, and classical, and identifying therapeutic vulnerabilities of these subtypes is an extremely high priority. We leveraged The Cancer Genome Atlas (TCGA) data, in particular for microRNA expression, to seek druggable core pathways in GBM. The E2F1-regulated miR-17~92 cluster and its analogs are shown to be highly expressed in proneural GBM and in GSC lines, suggesting the E2F cell cycle pathway might be a key driver in proneural GBM. Consistently, CDK4/6 inhibition with palbociclib preferentially inhibited cell proliferation in vitro in a majority of proneural GSCs versus those of other subtypes. Palbociclib treatment significantly prolonged survival of mice with established intracranial xenografts of a proneural GSC line. We show that most of these sensitive PN GSCs expressed higher levels of CDK6 and had intact Rb1, while two GSC lines with CDK4 overexpression and null Rb1 were highly resistant to palbociclib. Importantly, palbociclib treatment of proneural GSCs upregulated mesenchymal-associated markers and downregulated proneural-associated markers, suggesting that CDK4/6 inhibition induced proneural-mesenchymal transition and underscoring the enhanced role of the E2F cell cycle pathway in the proneural subtype. Lastly, the combination of palbociclib and N,N-diethylaminobenzaldehyde, an inhibitor of the mesenchymal driver ALDH1A3, showed strong synergistic inhibitory effects against proneural GSC proliferation. Taken together, our results reveal that proneural GBM has increased vulnerability to CDK4/6 inhibition, and the proneural subtype undergoes dynamic reprogramming upon palbociclib treatment-suggesting the need for a combination therapeutic strategy.


Cancer Research | 2016

Abstract 542: Assessing and augmenting the immune response to glioblastoma using repurposed pharmaceuticals

Breanna Brenneman; Desiree H. Floyd; Tajie H. Harris; Benjamin Purow

In order to improve the therapeutic responses to immunotherapies in glioblastoma (GBM) patients, a known pharmaceutical, ritanserin, was repurposed to augment cytotoxic T cell responses and suppress tumor growth. To determine the kinetics of the immune response to GBM, the transplantable syngeneic glioma cell line GL261 was used in C57BL/6 mice to assess the tumor immune response in the brain, spleen, and cervical lymph node via flow cytometry weekly for one month. After identifying the optimal T cell response temporally, mice with established tumors were treated with 100mg/kg of Ritanserin for one week and harvested post treatment. Ritanserin is able to inhibit the activity of diacylglycerol kinase alpha (DGKA), an enzyme that converts diacylglycerol to phosphatidic acid, and can inhibit tumor growth in mice. In addition, DGKA is expressed in T cells, and is more highly expressed in those that have entered an unresponsive (anergic) state, characteristic of dysfunctional T cell responses to tumors. To determine if ritanserin can counteract T cell anergy, T cell responses were determined by assessment of T cell number by subset (CD8+, CD4+, CD4+FoxP3+), by activation (CD62L-CD44+), and by tumor-specific response to ovalbumin-expressing GL261 tumor cells in the brain, spleen, and cervical lymph nodes. In comparison to mock-injected mice, tumor-bearing mice showed a robust T cell response to tumors in the brain, peaking at days 21 and 28 post injection. Treatment with ritanserin resulted in a trend toward increased number and activation of T cells in the brain compared to vehicle-treated mice. In addition, there was a significant increase in OVA-specific T cells in the brain with treatment. In conclusion, T cells respond robustly to brain tumors in mice, and ritanserin treatment appears to increase this response by increased activation and priming of tumor-specific T cells. This indicates that ritanserin may have combinatorial effects on T cells when combined with checkpoint inhibitors such as ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1). Future experiments will focus on combining ritanserin and other repurposed drugs with these checkpoint inhibitors and further characterizing the effect of ritanserin on T cells. Citation Format: Breanna R. Brenneman, Desiree H. Floyd, Tajie Harris, Benjamin Purow. Assessing and augmenting the immune response to glioblastoma using repurposed pharmaceuticals. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 542.


Cancer Research | 2011

Abstract 3316: An alpha-secretase inhibitor decreases glioma stem cell growth by inhibiting the Notch pathway and LMW Cyclin E

Desiree H. Floyd; Benjamin Kefas; Olexsandr Seleverstov; Charli Dominguez; Laurey Comeau; Benjamin Purow

The Notch pathway is deregulated in glioblastoma. Previous work suggests the inhibition of this pathway will have therapeutic benefits. The only inhibitors of Notch available block the gamma-secretase enzymatic complex necessary for processing Notch into the active form. However, Notch is also cleaved by alpha-secretase outside the plasma membrane, with the sheddases ADAM10 and 17. INCB3619 is a potent inhibitor of both ADAM10 and 17, and has been shown to inhibit growth of MCF-7 breast tumor cells when used with lapatinib. In this work, INCB3619 was used to inhibit endogenous Notch cleavage and downstream CBF-1 reporter activity in 0308 and 0822 human glioma stem cell lines. A microarray analysis of INCB3619 vs. DMSO treatment of 0308 cells identified many downregulated Notch pathway targets, but also identified new targets of INCB3619, such as CHI3L1/YKL40, an important prognostic indicator of many inflammatory diseases. Treatment of 0308 and 0822 glioma stem lines with INCB3619 inhibited their growth in culture due to a cell cycle arrest in G1 and a decrease in S phase cells. Growth inhibition by INCB3619 can be rescued with transfection of the active form of two Notch family members, NICD1 and 2. The growth inhibition of INCB3619 treated glioma stem cells is also stems from a shift in Cyclin E expression from a hyperactive low-molecular-weight form to the usual high-molecular-weight form. Also, INCB3619 induces higher expression of both p53 and p21, which can inhibit Cyclin E. Unlike with INCB3619, The treatment of glioma stem cells with DAPT, a well-known and potent gamma secretase inhibitor that cleaves Notch downstream of ADAM10 and 17, does not have a noticeable effect on Cyclin E processing or p21/p53 expression. INCB3619 therefore decreases glioma stem cell growth partly through a mechanism of Notch inhibition and also through inhibition of production of the hyperactive low-molecular-weight form of Cyclin E. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3316. doi:10.1158/1538-7445.AM2011-3316

Collaboration


Dive into the Desiree H. Floyd's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aizhen Xiao

University of Virginia

View shared research outputs
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

Jakub Godlewski

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge