Network


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

Hotspot


Dive into the research topics where Jessica A. Kilgore is active.

Publication


Featured researches published by Jessica A. Kilgore.


Nature Chemical Biology | 2009

Small molecule–mediated disruption of Wnt-dependent signaling in tissue regeneration and cancer

Baozhi Chen; Michael E. Dodge; Wei Tang; Jianming Lu; Zhiqiang Ma; Chih Wei Fan; Shuguang Wei; Wayne Hao; Jessica A. Kilgore; Noelle S. Williams; Michael G. Roth; James F. Amatruda; Chuo Chen; Lawrence Lum

SUMMARY The pervasive influence of secreted Wnt signaling proteins in tissue homeostasis and tumorigenesis has galvanized efforts to identify small molecules that target Wnt-mediated cellular responses. By screening a diverse synthetic chemical library, we have discovered two novel classes of small molecules that disrupt Wnt pathway responses - whereas one class inhibits the activity of Porcupine (Porcn), a membrane-bound acyltransferase that is essential to the production of Wnt proteins, the other abrogates destruction of Axin proteins, suppressors of Wnt/β-catenin pathway activity. With these small molecules we establish a chemical genetic approach for studying Wnt pathway responses and stem cell function in adult tissue. We achieve transient, reversible suppression of Wnt/β-catenin pathway response in vivo, and establish a mechanism-based approach to target cancerous cell growth. The signal transduction mechanisms shown here to be chemically tractable additionally contribute to Wnt-independent signal transduction pathways and thus could be broadly exploited for chemical genetics and therapeutic goals.


American Journal of Physiology-cell Physiology | 2011

Regulation of VEGF-induced endothelial cell migration by mitochondrial reactive oxygen species

Youxue Wang; Qun Zang; Zijuan Liu; Qian Wu; David L. Maass; Genevieve Dulan; Philip W. Shaul; Lisa Melito; Doug E. Frantz; Jessica A. Kilgore; Noelle S. Williams; Lance S. Terada; Fiemu E. Nwariaku

Endothelial migration is a crucial aspect of a variety of physiologic and pathologic conditions including atherosclerosis and vascular repair. Reactive oxygen species (ROS) function as second messengers during endothelial migration. Multiple intracellular sources of ROS are regulated by cellular context, external stimulus, and the microenvironment. However, the predominant source of ROS during endothelial cell (EC) migration and the mechanisms by which ROS regulate cell migration are incompletely understood. In this study, we tested the hypothesis that mitochondria-derived ROS (mtROS) regulate EC migration. In cultured human umbilical vein endothelial cells, VEGF increased mitochondrial metabolism, promoted mtROS production, and induced cell migration. Either the targeted mitochondrial delivery of the antioxidant, vitamin E (Mito-Vit-E), or the depletion of mitochondrial DNA abrogated VEGF-mediated mtROS production. Overexpression of mitochondrial catalase also inhibited VEGF-induced mitochondrial metabolism, Rac activation, and cell migration. Furthermore, these interventions suppressed VEGF-stimulated EC migration and blocked Rac1 activation in endothelial cells. Constitutively active Rac1 reversed Mito-Vit-E-induced inhibition of EC migration. Mito-Vit-E also attenuated carotid artery reendothelialization in vivo. These results provide strong evidence that mtROS regulate EC migration through Rac-1.


American Journal of Physiology-heart and Circulatory Physiology | 2012

Specific inhibition of mitochondrial oxidative stress suppresses inflammation and improves cardiac function in a rat pneumonia-related sepsis model

Qun Zang; Hesham A. Sadek; David L. Maass; Bobbie Martinez; Lisha Ma; Jessica A. Kilgore; Noelle S. Williams; Doug E. Frantz; Jane G. Wigginton; Fiemu E. Nwariaku; Steven E. Wolf; Joseph P. Minei

Using a mitochondria-targeted vitamin E (Mito-Vit-E) in a rat pneumonia-related sepsis model, we examined the role of mitochondrial reactive oxygen species in sepsis-mediated myocardial inflammation and subsequent cardiac contractile dysfunction. Sepsis was produced in adult male Sprague-Dawley rats via intratracheal injection of S. pneumonia (4 × 10(6) colony formation units per rat). A single dose of Mito-Vit-E, vitamin E, or control vehicle, at 21.5 μmol/kg, was administered 30 min postinoculation. Blood was collected, and heart tissue was harvested at various time points. Mito-Vit-E in vivo distribution was confirmed by mass spectrometry. In cardiac mitochondria, Mito-Vit-E improved total antioxidant capacity and suppressed H(2)O(2) generation, whereas vitamin E offered little effect. In cytosol, both antioxidants decreased H(2)O(2) levels, but only vitamin E strengthened antioxidant capacity. Mito-Vit-E protected mitochondrial structure and function in the heart during sepsis, demonstrated by reduction in lipid and protein oxidation, preservation of mitochondrial membrane integrity, and recovery of respiratory function. While both Mito-Vit-E and vitamin E suppressed sepsis-induced peripheral and myocardial production of proinflammatory cytokines (tumor necrosis factor-α, interleukin-1β, and interleukin-6), Mito-Vit-E exhibited significantly higher efficacy (P < 0.05). Stronger anti-inflammatory action of Mito-Vit-E was further shown by its near-complete inhibition of sepsis-induced myeloperoxidase accumulation in myocardium, suggesting its effect on neutrophil infiltration. Echocardiography analysis indicated that Mito-Vit-E ameliorated cardiac contractility of sepsis animals, shown by improved fractional shortening and ejection fraction. Together, our data suggest that targeted scavenging of mitochondrial reactive oxygen species protects mitochondrial function, attenuates tissue-level inflammation, and improves whole organ activities in the heart during sepsis.


Journal of Controlled Release | 2015

Esterase-activatable β-lapachone prodrug micelles for NQO1-targeted lung cancer therapy

Xinpeng Ma; Xiumei Huang; Zachary Moore; Gang Huang; Jessica A. Kilgore; Yiguang Wang; Suntrea T Hammer; Noelle S. Williams; David A. Boothman; Jinming Gao

UNLABELLED Lung cancer is one of the most lethal forms of cancer and current chemotherapeutic strategies lack broad specificity and efficacy. Recently, β-lapachone (β-lap) was shown to be highly efficacious in killing non-small cell lung cancer (NSCLC) cells regardless of their p53, cell cycle and caspase status. Pre-clinical and clinical use of β-lap (clinical form, ARQ501 or 761) is hampered by poor pharmacokinetics and toxicity due to hemolytic anemia. Here, we report the development and preclinical evaluation of β-lap prodrug nanotherapeutics consisting of diester derivatives of β-lap encapsulated in biocompatible and biodegradable poly(ethylene glycol)-b-poly(D,L-lactic acid) (PEG-b-PLA) micelles. Compared to the parent drug, diester derivatives of β-lap showed higher drug loading densities inside PEG-b-PLA micelles. After esterase treatment, micelle-delivered β-lap-dC3 and -dC6 prodrugs were converted to β-lap. Cytotoxicity assays using A549 and H596 lung cancer cells showed that both micelle formulations maintained NAD(P)H quinone oxidoreductase 1 (NQO1)-dependent cytotoxicity. However, antitumor efficacy study of β-lap-dC3 micelles against orthotopic A549 NSCLC xenograft-bearing mice showed significantly greater long-term survival over β-lap-dC6 micelles or β-lap-HPβCD complexes. Improved therapeutic efficacy of β-lap-dC3 micelles correlated with higher area under the concentration-time curves of β-lap in tumors, and enhanced pharmacodynamic endpoints (e.g., PARP1 hyperactivation, γH2AX, and ATP depletion). β-Lap-dC3 prodrug micelles provide a promising strategy for NQO1-targeted therapy of lung cancer with improved safety and antitumor efficacy.


Journal of Biological Chemistry | 2013

Identification of DNMT1 Selective Antagonists Using a Novel Scintillation Proximity Assay

Jessica A. Kilgore; Xinlin Du; Lisa Melito; Shuguang Wei; Changguang Wang; Hang Gyeong Chin; Bruce A. Posner; Sriharsa Pradhan; Joseph M. Ready; Noelle S. Williams

Background: DNA methylation contributes to the heritable regulation of gene expression. Results: Chemical inhibitors of DNA methyltransferase (DNMT) enzymes were identified. Conclusion: Several inhibitors nonspecifically inhibited multiple methyltransferases through generation of H2O2, but one compound showed selective inhibition of DNMT1 independent of the production of H2O2. Significance: Selective DNMT1 inhibitors will allow for more precise elucidation of the role of this enzyme in gene regulation. A novel scintillation proximity high throughput assay (SPA) to identify inhibitors of DNA methyltransferases was developed and used to screen over 180,000 compounds. The majority of the validated hits shared a quinone core and several were found to generate the reactive oxygen species, H2O2. Inhibition of the production of H2O2 by the addition of catalase blocked the ability of this group of compounds to inhibit DNA methyltransferase (DNMT) activity. However, a related compound, SW155246, was identified that existed in an already reduced form of the quinone. This compound did not generate H2O2, and catalase did not block its ability to inhibit DNA methyltransferase. SW155246 showed a 30-fold preference for inhibition of human DNMT1 versus human or murine DNMT3A or -3B, inhibited global methylation in HeLa cells, and reactivated expression of the tumor suppressor gene RASSF1A in A549 cells. To our knowledge, this work represents the first description of selective chemical inhibitors of the DNMT1 enzyme.


Cancer Cell | 2016

Leveraging an NQO1 Bioactivatable Drug for Tumor-Selective Use of Poly(ADP-ribose) Polymerase Inhibitors.

Xiumei Huang; Edward A. Motea; Zachary Moore; Jun Yao; Ying Dong; Gaurab Chakrabarti; Jessica A. Kilgore; Molly A. Silvers; Praveen L. Patidar; Agnieszka Cholka; Farjana Fattah; Yoonjeong Cha; Glenda G. Anderson; Rebecca Kusko; Michael Peyton; Jingsheng Yan; Xian Jin Xie; Venetia Sarode; Noelle S. Williams; John D. Minna; Muhammad Shaalan Beg; David E. Gerber; Erik A. Bey; David A. Boothman

Therapeutic drugs that block DNA repair, including poly(ADP-ribose) polymerase (PARP) inhibitors, fail due to lack of tumor-selectivity. When PARP inhibitors and β-lapachone are combined, synergistic antitumor activity results from sustained NAD(P)H levels that refuel NQO1-dependent futile redox drug recycling. Significant oxygen-consumption-rate/reactive oxygen species cause dramatic DNA lesion increases that are not repaired due to PARP inhibition. In NQO1+ cancers, such as non-small-cell lung, pancreatic, and breast cancers, cell death mechanism switches from PARP1 hyperactivation-mediated programmed necrosis with β-lapachone monotherapy to synergistic tumor-selective, caspase-dependent apoptosis with PARP inhibitors and β-lapachone. Synergistic antitumor efficacy and prolonged survival were noted in human orthotopic pancreatic and non-small-cell lung xenograft models, expanding use and efficacy of PARP inhibitors for human cancer therapy.


Archive | 2016

NQO1 Bioactivatable Drugs Enhance Radiation Responses

Erik A. Bey; Julia C. Meade; Molly A. Silvers; Edward A. Motea; Praveen L. Patidar; Rolf A. Brekken; Stanislaw Deja; Matthew E. Merritt; Jessica A. Kilgore; Yuliang Liu; Xiumei Huang; Longshan Li; John S. Yordy; Noelle S. Williams; Jinming Gao; David A. Boothman

Inhibitors of cancer-specific pathways can selectively kill off tumor cells. However, heterogeneity of neoplastic tissue often allows other cancer cells to repopulate the tissue area, leading to regeneration of resistant disease. β-Lapachone is a novel bioactivatable drug that relies specifically on tumor-directed upregulated levels of NAD(P)H:quinone oxidoreductase 1 (NQO1) to kill most solid cancers, such as 90 % of pancreatic and non-small cell lung, 60 % of breast, colon, and prostate, as well as 50 % of head and neck cancers. Once β-lapachone is bioactivated by the NQO1 enzyme, massive levels of hydrogen peroxide are produced that, in turn, damage the DNA of cancer cells, while associated normal tissues, which lack NQO1, are protected by high levels of catalase. If tumors are irradiated prior to applying β-lapachone, the drug (clinical form, ARQ761) can work in combination with the vast spectrum of DNA lesions created by ionizing radiation, particularly DNA base lesions, single and double strand breaks (SSBs and DSBs), in addition to the massive hydrogen peroxide-based lesions created by β-lapachone, to cause tumor-dependent poly(ADP-ribose) polymerase 1 (PARP1) hyperactivation. Once tumor-selective PARP hyperactivation is induced in cancer cells, they die due to low concomitant catalase levels. In contrast, associated normal tissue, as well as other normal tissue, lack elevated levels of NQO1 and have high catalase levels. Cancer cell death ultimately occurs by NAD+-depletion, where resistance to NQO1 bioactivatable drugs has not been noted to date. Current studies are focused on pancreatic and non-small cell lung cancers, as NQO1 is elevated in nearly all of these cancers.


Cancer Research | 2012

An NQO1 Substrate with Potent Antitumor Activity That Selectively Kills by PARP1-Induced Programmed Necrosis

Xiumei Huang; Ying Dong; Erik A. Bey; Jessica A. Kilgore; Joseph S. Bair; Long Shan Li; Malina Patel; Elizabeth I. Parkinson; Yiguang Wang; Noelle S. Williams; Jinming Gao; Paul J. Hergenrother; David A. Boothman


Archive | 2016

by mitochondrial reactive oxygen species Regulation of VEGF-induced endothelial cell migration

Lance S. Terada; Fiemu E. Nwariaku; Philip W. Shaul; Lisa Melito; Doug E. Frantz; Jessica A. Kilgore; Noelle S. Williams; Youxue Wang; Qun Zang; Zijuan Liu; Qian Wu; David L. Maass; Genevieve Dulan


Archive | 2015

cardiac function in a rat pneumonia-related sepsis model Specific inhibition of mitochondrial oxidative stress suppresses inflammation and improves

Leigh A. Callahan; Gerald S. Supinski; G. S. Supinski; Michael P. Murphy; Leigh Ann Callahan; Joseph P. Minei; Noelle S. Williams; Doug E. Frantz; Jane G. Wigginton; Fiemu E. Nwariaku; Steven E. Wolf; Qun Zang; Hesham A. Sadek; David L. Maass; Bobbie Martinez; Lisha Ma; Jessica A. Kilgore

Collaboration


Dive into the Jessica A. Kilgore's collaboration.

Top Co-Authors

Avatar

Noelle S. Williams

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

David L. Maass

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Doug E. Frantz

University of Texas at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Lisa Melito

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Qun Zang

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

David A. Boothman

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Fiemu E. Nwariaku

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Xiumei Huang

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Erik A. Bey

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jinming Gao

University of Texas Southwestern Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge