Previous Page  5 / 15 Next Page
Information
Show Menu
Previous Page 5 / 15 Next Page
Page Background

J u n e 1 1 - 1 3 , 2 0 1 8 | D u b l i n , I r e l a n d

allied

academies

Page 69

CANCER STEM CELLS AND

ONCOLOGY RESEARCH

11

th

International Conference on

Journal of Medical Oncology and Therapeutics

|

Volume 3

Seamus Caragher et al., J Med Oncl Ther 2018, Volume 3

ACTIVATION OF DOPAMINE RECEPTOR

2 INCREASES TUMORGENECITY

AND ALTERS METABOLISM IN

GLIOBLASTOMA IN SUB-TYPE

DEPENDENT MANNER

Seamus Caragher, Jack Shireman, Cheol Hong Park, Mei

Huang, Jason Miska, Herbert Meltzer, Maijic Lesniak

and

Atique Ahmed

Northwestern University, USA

O

f all cancers, glioblastoma (GBM) remains one of the least treatable.

Evidence indicates cellular plasticity—GBM cells’ ability to adopt

various expression profiles and functional attributes—is a key factor in this

aggressive phenotype. This process includes the ability of differentiated

GBM cells to attain a glioma-intiating cell (GIC) phenotype, characterized

by heightened therapy-resistance and elevated self-renewal capacity. A

variety of factors in the microenvironment have been shown to influence

this process, including hypoxia, acidity, and therapeutic stress. Therefore,

a better understanding of the mechanisms governing this conversion

is needed. Developmental neurobiology suggests that dopamine, a

monoamine neurotransmitter, may represent one such factor. Dopamine

signaling influences differentiation of brain progenitor cells, highly similar

to GICs. We set out to investigate how dopamine influences cellular

plasticity in GBM. First, we analyzed epigenetic regulation of the five

dopamine receptors (DRDs) in patient derived xenograft (PDX) cells. We

found that therapy induces increased acetylation of H3K27 in the DRD2

promoter. Western blots and FACS confirmed increased DRD2 protein.

Next, we performed neurosphere assays in the presence of a specific

DRD2 agonist. Agonist treated classical PDX cells increased in sphere-

forming capacity, while proneural PDX showed no change. Blocking DRD2

attenuated neurosphere-formation. To determine what pathways drive

this DRD2 activated plasticity, we performed bioinformatics analysis

of human GBM samples. DRD2 expression correlated positively with

hypoxia inducible factor (HIF) signaling. Agonist treatment of PDX cells

induced HIF protein, despite normoxic conditions. Microarray analysis of

HIF genes confirmed subtype-dependent alterations in gene expression

following DRD2 activation. Finally, we examined how these gene

expression changes influence metabolism, a key functional output of

HIF signaling. Seahorse analysis revealed classical GBM cells augment

glycolytic rate following DRD2 activation, while proneural GBM cells

decrease their consumption of glucose. In summary, these data highlight

the contribution of CNS-specific molecules to cellular plasticity of GBM

Seamus Caragher earned his B.S. in Neurobiolo-

gy, summa cum laude and Phi Beta Kappa, from

Georgetown University in 2016. He then worked

in the laboratory of Atique Ahmed, PhD at the

Lurie Cancer Center of Northwestern University,

focusing on cellular plasticity and the influence

of the brain microenvironment in glioblastoma.

He is currently pursuing an MSc in Cancer Sci-

ences at the University of Glasgow as a British

Marshall Scholar.

seamuscaragher@gmail.com

BIOGRAPHY

cells and provide evidence for functional

differences in genetically defined tumor

subpopulations.