Network


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

Hotspot


Dive into the research topics where Jason Chesney is active.

Publication


Featured researches published by Jason Chesney.


Breast Cancer Research | 2008

Targeting aspartate aminotransferase in breast cancer

Joshua Thornburg; Kristin Nelson; Brian Clem; Andrew N. Lane; Sengodagounder Arumugam; Allan Simmons; John W. Eaton; Sucheta Telang; Jason Chesney

IntroductionGlycolysis is increased in breast adenocarcinoma cells relative to adjacent normal cells in order to produce the ATP and anabolic precursors required for survival, growth and invasion. Glycolysis also serves as a key source of the reduced form of cytoplasmic nicotinamide adenine dinucleotide (NADH) necessary for the shuttling of electrons into mitochondria for electron transport. Lactate dehydrogenase (LDH) regulates glycolytic flux by converting pyruvate to lactate and has been found to be highly expressed in breast tumours. Aspartate aminotransferase (AAT) functions in tandem with malate dehydrogenase to transfer electrons from NADH across the inner mitochondrial membrane. Oxamate is an inhibitor of both LDH and AAT, and we hypothesised that oxamate may disrupt the metabolism and growth of breast adenocarcinoma cells.MethodsWe examined the effects of oxamate and the AAT inhibitor amino oxyacetate (AOA) on 13C-glucose utilisation, oxygen consumption, NADH and ATP in MDA-MB-231 cells. We then determined the effects of oxamate and AOA on normal human mammary epithelial cells and MDA-MB-231 breast adenocarcinoma cell proliferation, and on the growth of MDA-MB-231 cells as tumours in athymic BALB/c female mice. We ectopically expressed AAT in MDA-MB-231 cells and examined the consequences on the cytostatic effects of oxamate. Finally, we examined the effect of AAT-specific siRNA transfection on MDA-MB-231 cell proliferation.ResultsWe found that oxamate did not attenuate cellular lactate production as predicted by its LDH inhibitory activity, but did have an anti-metabolic effect that was similar to AAT inhibition with AOA. Specifically, we found that oxamate and AOA decreased the flux of 13C-glucose-derived carbons into glutamate and uridine, both products of the mitochondrial tricarboxylic acid cycle, as well as oxygen consumption, a measure of electron transport chain activity. Oxamate and AOA also selectively suppressed the proliferation of MDA-MB-231 cells relative to normal human mammary epithelial cells and decreased the growth of MDA-MB-231 breast tumours in athymic mice. Importantly, we found that ectopic expression of AAT in MDA-MB-231 cells conferred resistance to the anti-proliferative effects of oxamate and that siRNA silencing of AAT decreased MDA-MB-231 cell proliferation.ConclusionsWe conclude that AAT may be a valid molecular target for the development of anti-neoplastic agents.


Journal of Translational Medicine | 2008

Transient T cell depletion causes regression of melanoma metastases

Mary Ann Rasku; Amy Clem; Sucheta Telang; Beverly Taft; Kelly Gettings; Hana Gragg; Daniel W. Cramer; Sheron C. Lear; Kelly M. McMasters; Donald M. Miller; Jason Chesney

BackgroundCognate immunity against neoplastic cells depends on a balance between effector T cells and regulatory T (Treg) cells. Treg cells prevent immune attack against normal and neoplastic cells by directly suppressing the activation of effector CD4+ and CD8+ T cells. We postulated that a recombinant interleukin 2/diphtheria toxin conjugate (DAB/IL2; Denileukin Diftitox; Ontak) may serve as a useful strategy to deplete Treg cells and break tolerance against neoplastic tumors in humans.MethodsWe administered DAB/IL2 (12 μg/kg; four daily doses; 21 day cycles) to 16 patients with metastatic melanoma and measured the effects on the peripheral blood concentration of several T cell subsets and on tumor burden.ResultsWe found that DAB/IL2 caused a transient depletion of Treg cells as well as total CD4+ and CD8+ T cells (< 21 days). T cell repopulation coincided with the de novo appearance of melanoma antigen-specific CD8+ T cells in several patients as determined by flow cytometry using tetrameric MART-1, tyrosinase and gp100 peptide/MHC conjugates. Sixteen patients received at least one cycle of DAB/IL2 and five of these patients experienced regressions of melanoma metastases as measured by CT and/or PET imaging. One patient experienced a near complete response with the regression of several hepatic and pulmonary metastases coupled to the de novo appearance of MART-1-specific CD8+ T cells. A single metastatic tumor remained in this patient and, after surgical resection, immunohistochemical analysis revealed MART1+ melanoma cells surrounded by CD8+ T cells.ConclusionTaken together, these data indicate that transient depletion of T cells in cancer patients may disrupt the homeostatic control of cognate immunity and allow for the expansion of effector T cells with specificity against neoplastic cells. Several T cell depleting agents are clinically available and this study provides strong rationale for an examination of their efficacy in cancer patients.Trial registrationNCT00299689 (ClinicalTrials.gov Identifier).


Current Opinion in Clinical Nutrition and Metabolic Care | 2006

6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase and tumor cell glycolysis.

Jason Chesney

Purpose of reviewNeoplastic cells metabolize abundant glucose relative to normal cells in order to satisfy the increased energetic and anabolic needs of the transformed state. This review will summarize the requirement of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases for the regulation of glycolysis in cancer cells and their potential utility as targets for the development of antineoplastic agents. Recent findingsThe steady-state concentration of fructose-2,6-bisphosphate controls the overall rate of glycolysis by allosterically activating a rate-limiting enzyme, 6-phosphofructo-1-kinase. The intracellular concentration of fructose-2,6-bisphosphate is controlled by a family of bifunctional 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases that are encoded by four independent genes (PFKFB1–4). The 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase encoded by the PFKFB3 gene has the highest kinase:phosphatase activity ratio of the four enzymes and thus contributes significantly to the synthesis of fructose-2,6-bisphosphate. PFKFB3 is activated by mitogenic, inflammatory and hypoxic stimuli, and was recently found to be constitutively expressed by several human leukemias and solid tumor cells. By setting the intracellular fructose-2,6-bisphosphate concentration, PFKFB3 controls glycolytic flux to lactate and the nonoxidative pentose shunt, and is selectively required for the tumorigenic growth of ras-transformed cells. SummaryThese findings demonstrate a key role for the 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases in neoplastic transformation and provide rationale for the development of agents that selectively inhibit the PFKFB3 enzyme as antineoplastic agents.


Journal of Translational Medicine | 2012

Small molecule inhibition of 6-phosphofructo-2-kinase suppresses t cell activation.

Sucheta Telang; Brian Clem; Alden C. Klarer; Amy Clem; John O. Trent; Richard Bucala; Jason Chesney

BackgroundT cell activation is associated with a rapid increase in intracellular fructose-2,6-bisphosphate (F2,6BP), an allosteric activator of the glycolytic enzyme, 6-phosphofructo-1-kinase. The steady state concentration of F2,6BP in T cells is dependent on the expression of the bifunctional 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases (PFKFB1-4) and the fructose-2,6-bisphosphatase, TIGAR. Of the PFKFB family of enzymes, PFKFB3 has the highest kinase:bisphosphatase ratio and has been demonstrated to be required for T cell proliferation. A small molecule antagonist of PFKFB3, 3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO), recently has been shown to reduce F2,6BP synthesis, glucose uptake and proliferation in transformed cells. We hypothesized that the induction of PFKFB3 expression may be required for the stimulation of glycolysis in T cells and that exposure to the PFKFB3 antagonist, 3PO, would suppress T cell activation.MethodsWe examined PFKFB1-4 and TIGAR expression and F2,6BP concentration in purified CD3+ T cells stimulated with microbead-conjugated agonist antibodies specific for CD3 and the co-stimulatory receptor, CD28. We then determined the effect of 3PO on anti-CD3/anti-CD28-induced T cell activation, F2,6BP synthesis, 2-[1-14C]-deoxy-d-glucose uptake, lactate secretion, TNF-α secretion and proliferation. Finally, we examined the effect of 3PO administration on the development of delayed type hypersensitivity to methylated BSA and on imiquimod-induced psoriasis in mice.ResultsWe found that purified human CD3+ T cells express PFKFB2, PFKFB3, PFKFB4 and TIGAR, and that anti-CD3/anti-CD28 conjugated microbeads stimulated a >20-fold increase in F2,6BP with a coincident increase in protein expression of the PFKFB3 family member and a decrease in TIGAR protein expression. We then found that exposure to the PFKFB3 small molecule antagonist, 3PO (1–10 μM), markedly attenuated the stimulation of F2,6BP synthesis, 2-[1-14C]-deoxy-D-glucose uptake, lactate secretion, TNF-α secretion and T cell aggregation and proliferation. We examined the in vivo effect of 3PO on the development of delayed type hypersensitivity to methylated BSA and on imiquimod-induced psoriasis in mice and found that 3PO suppressed the development of both T cell-dependent models of immunity in vivo.ConclusionsOur data demonstrate that inhibition of the PFKFB3 kinase activity attenuates the activation of T cells in vitro and suppresses T cell dependent immunity in vivo and indicate that small molecule antagonists of PFKFB3 may prove effective as T cell immunosuppressive agents.


Journal of Biological Chemistry | 2014

Estradiol stimulates glucose metabolism via 6-phosphofructo-2-kinase (PFKFB3)

Yoannis Imbert-Fernandez; Brian Clem; Julie O'Neal; Daniel Alan Kerr; Robert Spaulding; Lilibeth Lanceta; Amy Clem; Sucheta Telang; Jason Chesney

Background: The regulation of glucose metabolism by estradiol is poorly defined. Results: We find that estradiol stimulates glucose metabolism in part by stimulating the production of fructose 2,6-bisphosphate by PFKFB3. Conclusion: PFKFB3 is a downstream target of estradiol required to stimulate glucose metabolism. Significance: Combined targeting of PFKFB3 and the estrogen receptor may prove beneficial to ER+ stage IV breast cancer patients. Estradiol (E2) administered to estrogen receptor-positive (ER+) breast cancer patients stimulates glucose uptake by tumors. Importantly, this E2-induced metabolic flare is predictive of the clinical effectiveness of anti-estrogens and, as a result, downstream metabolic regulators of E2 are expected to have utility as targets for the development of anti-breast cancer agents. The family of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases (PFKFB1–4) control glycolytic flux via their product, fructose-2,6-bisphosphate (F26BP), which activates 6-phosphofructo-1-kinase (PFK-1). We postulated that E2 might promote PFKFB3 expression, resulting in increased F26BP and glucose uptake. We demonstrate that PFKFB3 expression is highest in stage III lymph node metastases relative to normal breast tissues and that exposure of human MCF-7 breast cancer cells to E2 causes a rapid increase in [14C]glucose uptake and glycolysis that is coincident with an induction of PFKFB3 mRNA (via ER binding to its promoter), protein expression and the intracellular concentration of its product, F26BP. Importantly, selective inhibition of PFKFB3 expression and activity using siRNA or a PFKFB3 inhibitor markedly reduces the E2-mediated increase in F26BP, [14C]glucose uptake, and glycolysis. Furthermore, co-treatment of MCF-7 cells with the PFKFB3 inhibitor and the anti-estrogen ICI 182,780 synergistically induces apoptotic cell death. These findings demonstrate for the first time that the estrogen receptor directly promotes PFKFB3 mRNA transcription which, in turn, is required for the glucose metabolism and survival of breast cancer cells. Importantly, these results provide essential preclinical information that may allow for the ultimate design of combinatorial trials of PFKFB3 antagonists with anti-estrogen therapies in ER+ stage IV breast cancer patients.


The American Journal of the Medical Sciences | 2009

Digitalis, A Targeted Therapy for Cancer?

M. Iltaf Khan; Jason Chesney; Damian A. Laber; Donald M. Miller

The clinical benefit of digitalis for patients with heart disease is well established. However, recent studies have also suggested that digitalis has antineoplastic activities at clinically relevant serum concentrations. Much of the early evidence supporting the anticancer activity of digitalis has been circumstantial. Observational studies suggest a protective benefit and improved outcomes in patients who develop cancer while they are taking digitalis. The mechanism by which digitalis selectively affects the growth of malignant cells is complex, involving several important signaling pathways. Experiments to determine its mechanism of action have demonstrated that digitalis inhibits cell growth and angiogenesis and induces apoptosis in multiple cancer cell lines. Most, if not all, of these effects are mediated through its target enzyme, sodium- and potassium-activated adenosine triphosphatase. This article reviews the literature, which supports the use of digitalis in patients with malignancies with a discussion of the potential mechanisms of action. We hypothesize that sodium- and potassium-activated adenosine triphosphatase is an important new target for cancer therapy. It is reasonable to expect that the addition of digitalis to current cancer treatments will improve the clinical outcomes.


Nature Communications | 2013

Rb1 family mutation is sufficient for sarcoma initiation

Yongqing Liu; Ester Sánchez-Tilló; Xiaoqin Lu; Brian Clem; Sucheta Telang; Alfred B. Jenson; Miriam Cuatrecasas; Jason Chesney; Antonio Postigo; Douglas C. Dean

It is thought that genomic instability precipitated by Rb1 pathway loss rapidly triggers additional cancer gene mutations, accounting for rapid tumour onset following Rb1 mutation. However, recent whole-genome sequencing of retinoblastomas demonstrated little genomic instability, but instead suggested rapid epigenetic activation of cancer genes. These results raise the possibility that loss of the Rb1 pathway, which is a hallmark of cancers, might be sufficient for cancer initiation. Yet, mutation of the Rb1 family or inactivation of the Rb1 pathway in primary cells has proven insufficient for tumour initiation. Here we demonstrate that traditional nude mouse assays impose an artificial anoikis and proliferation barrier that prevents Rb1 family mutant fibroblasts from initiating tumours. By circumventing this barrier, we show that primary fibroblasts with only an Rb1 family mutation efficiently form sarcomas in nude mice, and a Ras-ZEB1-Akt pathway then causes transition of these tumours to an invasive phenotype.


Cancer and Metabolism | 2014

Fructose-2,6-bisphosphate links glucose to survival and growth

Abdullah Yalcin; Yoannis Imbert-Fernandez; Amy Clem; Julie O’Neal; Sucheta Telang; Brian Clem; Jason Chesney

Background Glucose starvation, as occurs in neoplastic tumors, causes G1 arrest and/or apoptosis. Although the mechanisms for these events are poorly understood, reductions in nucleotide synthesis and ATP are widely believed to restrict progression into the S phase and cause apoptosis, respectively. Recently, a side product of glycolysis and stimulator of PFK-1, fructose 2,6-bisphosphate (F2,6BP), was found to be synthesized in the nucleus of cells by the enzyme PFKFB3 and to stimulate proliferation in part by activating cyclin dependent kinases (Cdk). We hypothesized that glucose deprivation would reduce F2,6BP which in turn would suppress the activity of Cdks and cause G1 arrest and apoptosis.


Cancer Research | 2009

Inhibition of 6-phosphofructo-2-kinase suppresses breast tumor growth in vivo.

Brian Clem; Sucheta Telang; Amy Clem; Umesh Goswami; Jason Chesney

Abstract #3064 Phosphofructo-1-kinase, a rate-limiting enzyme of glycolysis, is activated in neoplastic cells by fructose-2,6-bisphosphate (Fru-2,6-BP), a product of four 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase isozymes (PFKFB1-4). The inducible PFKFB3 isozyme is constitutively expressed by neoplastic cells and required for the high glycolytic rate and anchorage-independent growth of ras -transformed cells. We report that siRNA silencing or small molecule inhibition (3PO) of PFKFB3 causes a decrease in cell proliferation and metabolic flux in several breast cancer cell lines (MDA-MB-231, MDA-MB-468, BT-549, BT-474, SK-BR-3, ZR-75-1, MCF-7, and MCF-10A). Furthermore, 0.075mg/g 3PO administered via intraperitoneal injection suppresses tumor growth in two transgenic mouse models of breast cancer (MMTV- H-ras : 30% inhibition; MMTV- Erbb2(Her2/neu ): 60% inhibition). Interestingly, the Her2 positive cell lines (SK-BR-3, BT-549) and the Her2 mouse transgenic model were more susceptible to PFKFB3 inhibition suggesting that the Her2 phenotype may be more sensitive to disruption of glycolytic flux. Taken together, these data support the clinical development of 3PO and other PFKFB3 inhibitors as chemotherapeutic agents against breast cancer. Citation Information: Cancer Res 2009;69(2 Suppl):Abstract nr 3064.


Cell | 2013

Role of PFKFB3-Driven Glycolysis in Vessel Sprouting

Katrien De Bock; Maria Georgiadou; Sandra Schoors; Anna Kuchnio; Brian W. Wong; Anna Rita Cantelmo; Annelies Quaegebeur; Bart Ghesquière; Sandra Cauwenberghs; Guy Eelen; Li-Kun Phng; Inge Betz; Bieke Tembuyser; Katleen Brepoels; Jonathan Welti; Ilse Geudens; Inmaculada Segura; Bert Cruys; Franscesco Bifari; Raquel Blanco; Sabine Wyns; Jeroen Vangindertael; Susana Rocha; Russel T Collins; Sebastian Munck; Dirk Daelemans; Hiromi Imamura; Roland Devlieger; Mark H. Rider; Paul P. Van Veldhoven

Collaboration


Dive into the Jason Chesney's collaboration.

Top Co-Authors

Avatar

Sucheta Telang

University of Louisville

View shared research outputs
Top Co-Authors

Avatar

Amy Clem

University of Louisville

View shared research outputs
Top Co-Authors

Avatar

Brian Clem

University of Louisville

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Beverly Taft

University of Louisville

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hana Gragg

University of Louisville

View shared research outputs
Top Co-Authors

Avatar

Julie O’Neal

University of Louisville

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge