Network


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

Hotspot


Dive into the research topics where Jillian Onufrak is active.

Publication


Featured researches published by Jillian Onufrak.


PLOS ONE | 2014

Cardiovascular Risks Associated with Low Dose Ionizing Particle Radiation

Xinhua Yan; Sharath P. Sasi; Hannah Gee; Juyong Lee; Yongyao Yang; Raman Mehrzad; Jillian Onufrak; Jin Song; Heiko Enderling; Akhil Agarwal; Layla Rahimi; James P. Morgan; Paul F. Wilson; Joseph P. Carrozza; Kenneth Walsh; Raj Kishore; David A. Goukassian

Previous epidemiologic data demonstrate that cardiovascular (CV) morbidity and mortality may occur decades after ionizing radiation exposure. With increased use of proton and carbon ion radiotherapy and concerns about space radiation exposures to astronauts on future long-duration exploration-type missions, the long-term effects and risks of low-dose charged particle irradiation on the CV system must be better appreciated. Here we report on the long-term effects of whole-body proton (1H; 0.5 Gy, 1 GeV) and iron ion (56Fe; 0.15 Gy, 1GeV/nucleon) irradiation with and without an acute myocardial ischemia (AMI) event in mice. We show that cardiac function of proton-irradiated mice initially improves at 1 month but declines by 10 months post-irradiation. In AMI-induced mice, prior proton irradiation improved cardiac function restoration and enhanced cardiac remodeling. This was associated with increased pro-survival gene expression in cardiac tissues. In contrast, cardiac function was significantly declined in 56Fe ion-irradiated mice at 1 and 3 months but recovered at 10 months. In addition, 56Fe ion-irradiation led to poorer cardiac function and more adverse remodeling in AMI-induced mice, and was associated with decreased angiogenesis and pro-survival factors in cardiac tissues at any time point examined up to 10 months. This is the first study reporting CV effects following low dose proton and iron ion irradiation during normal aging and post-AMI. Understanding the biological effects of charged particle radiation qualities on the CV system is necessary both for the mitigation of space exploration CV risks and for understanding of long-term CV effects following charged particle radiotherapy.


American Journal of Physiology-heart and Circulatory Physiology | 2015

Low-dose radiation affects cardiac physiology: gene networks and molecular signaling in cardiomyocytes

Matthew A. Coleman; Sharath P. Sasi; Jillian Onufrak; Mohan Natarajan; Krishnan Manickam; John Schwab; Sujatha Muralidharan; Leif E. Peterson; Yuriy O. Alekseyev; Xinhua Yan; David A. Goukassian

There are 160,000 cancer patients worldwide treated with particle radiotherapy (RT). With the advent of proton, and high (H) charge (Z) and energy (E) HZE ionizing particle RT, the cardiovascular diseases risk estimates are uncertain. In addition, future deep space exploratory-type missions will expose humans to unknown but low doses of particle irradiation (IR). We examined molecular responses using transcriptome profiling in left ventricular murine cardiomyocytes isolated from mice that were exposed to 90 cGy, 1 GeV proton ((1)H) and 15 cGy, 1 GeV/nucleon iron ((56)Fe) over 28 days after exposure. Unsupervised clustering analysis of gene expression segregated samples according to the IR response and time after exposure, with (56)Fe-IR showing the greatest level of gene modulation. (1)H-IR showed little differential transcript modulation. Network analysis categorized the major differentially expressed genes into cell cycle, oxidative responses, and transcriptional regulation functional groups. Transcriptional networks identified key nodes regulating expression. Validation of the signal transduction network by protein analysis and gel shift assay showed that particle IR clearly regulates a long-lived signaling mechanism for ERK1/2, p38 MAPK signaling and identified NFATc4, GATA4, STAT3, and NF-κB as regulators of the response at specific time points. These data suggest that the molecular responses and gene expression to (56)Fe-IR in cardiomyocytes are unique and long-lasting. Our study may have significant implications for the efforts of National Aeronautics and Space Administration to develop heart disease risk estimates for astronauts and for patients receiving conventional and particle RT via identification of specific HZE-IR molecular markers.


Stem Cells International | 2015

Particle Radiation-Induced Nontargeted Effects in Bone-Marrow-Derived Endothelial Progenitor Cells

Sharath P. Sasi; Daniel Park; Sujatha Muralidharan; Justin Wage; Albert Kiladjian; Jillian Onufrak; Heiko Enderling; Xinhua Yan; David A. Goukassian

Bone-marrow- (BM-) derived endothelial progenitor cells (EPCs) are critical for endothelial cell maintenance and repair. During future space exploration missions astronauts will be exposed to space irradiation (IR) composed of a spectrum of low-fluence protons (1H) and high charge and energy (HZE) nuclei (e.g., iron-56Fe) for extended time. How the space-type IR affects BM-EPCs is limited. In media transfer experiments in vitro we studied nontargeted effects induced by 1H- and 56Fe-IR conditioned medium (CM), which showed significant increase in the number of p-H2AX foci in nonirradiated EPCs between 2 and 24 h. A 2–15-fold increase in the levels of various cytokines and chemokines was observed in both types of IR-CM at 24 h. Ex vivo analysis of BM-EPCs from single, low-dose, full-body 1H- and 56Fe-IR mice demonstrated a cyclical (early 5–24 h and delayed 28 days) increase in apoptosis. This early increase in BM-EPC apoptosis may be the effect of direct IR exposure, whereas late increase in apoptosis could be a result of nontargeted effects (NTE) in the cells that were not traversed by IR directly. Identifying the role of specific cytokines responsible for IR-induced NTE and inhibiting such NTE may prevent long-term and cyclical loss of stem and progenitors cells in the BM milieu.


Radiation Research | 2017

Different Sequences of Fractionated Low-Dose Proton and Single Iron-Radiation-Induced Divergent Biological Responses in the Heart

Sharath P. Sasi; Xinhua Yan; Marian Zuriaga-Herrero; Hannah Gee; Juyong Lee; Raman Mehrzad; Jin Song; Jillian Onufrak; James P. Morgan; Heiko Enderling; Kenneth Walsh; Raj Kishore; David A. Goukassian

Deep-space travel presents risks of exposure to ionizing radiation composed of a spectrum of low-fluence protons (1H) and high-charge and energy (HZE) iron nuclei (e.g., 56Fe). When exposed to galactic cosmic rays, each cell in the body may be traversed by 1H every 3–4 days and HZE nuclei every 3–4 months. The effects of low-dose sequential fractionated 1H or HZE on the heart are unknown. In this animal model of simulated ionizing radiation, middle-aged (8–9 months old) male C57BL/6NT mice were exposed to radiation as follows: group 1, nonirradiated controls; group 2, three fractionated doses of 17 cGy 1H every other day (1H × 3); group 3, three fractionated doses of 17 cGy 1H every other day followed by a single low dose of 15 cGy 56Fe two days after the final 1H dose (1H × 3 + 56Fe); and group 4, a single low dose of 15 cGy 56Fe followed (after 2 days) by three fractionated doses of 17 cGy 1H every other day (56Fe + 1H × 3). A subgroup of mice from each group underwent myocardial infarction (MI) surgery at 28 days postirradiation. Cardiac structure and function were assessed in all animals at days 7, 14 and 28 after MI surgery was performed. Compared to the control animals, the treatments that groups 2 and 3 received did not induce negative effects on cardiac function or structure. However, compared to all other groups, the animals in group 4, showed depressed left ventricular (LV) functions at 1 month with concomitant enhancement in cardiac fibrosis and induction of cardiac hypertrophy signaling at 3 months. In the irradiated and MI surgery groups compared to the control group, the treatments received by groups 2 and 4 did not induce negative effects at 1 month postirradiation and MI surgery. However, in group 3 after MI surgery, there was a 24% increase in mortality, significant decreases in LV function and a 35% increase in post-infarction size. These changes were associated with significant decreases in the angiogenic and cell survival signaling pathways. These data suggest that fractionated doses of radiation induces cellular and molecular changes that result in depressed heart functions both under basal conditions and particularly after myocardial infarction.


Journal of Cardiovascular Diseases and Diagnosis | 2013

Neuregulin1 Improved Cardiac Function in Doxorubicin-Treated Mice with Cardiomyocyte-Specific over expression of a Dominant-NegativePI3Kp110ñ

Yun Bian; Marcy Silver; Jillian Onufrak; Kalon K.L. Ho; Mark A. Marchionni; Peter M. Kang; David A. Goukassian; Joseph P. Carrozza; James P. Morgan; Xinhua Yan

Neuregulin1s (NRG1s) are effective for protecting the heart from various stresses. Our previous studies show that heterozygous knockout of the NRG1 gene worsens while injections of a recombinant NRG1 improve cardiac function in Chemotherapy Drug Doxorubicin (DOX) - treated mice. In cultured cardiomyocytes, studies show that the Phosphoinositide 3-Kinase (PI3K) pathway is necessary for NRG1 to reduce DOX-induced apoptosis and loss of cardiac troponins. Here, we test whether PI3Kp110α, a Class IA PI3K isoform, is necessary for NRG1’s cardioprotective effects in DOX-treated mice. DOX was administered to mice with cardiomyocyte-specific overexpression of a constitutively active PI3Kp110α (CaPI3K), or a dominant negative PI3Kp110α (dnPI3K). Solvent or NRG1 was administered concurrently with DOX to dnPI3K mice. Our results showed that survival and cardiac function were improved in CaPI3K, but worsened in dnPI3K mice. Concurrent NRG1 injections significantly improved survival and cardiac function in dnPI3K mice, which were associated with increased activation of ERK1/2 and mTORC1 in the heart. These results have demonstrated that PI3Kp110α is important for protecting the heart from DOX. Further, the results suggest that PI3Kp110α is not necessary for NRG1 to protect the heart from DOX. NRG1 may protect the heart by activating alternative survival pathways, such as ERK1/2 and mTORC1, in DOX-treated dnPI3K mouse hearts.


Stem Cells International | 2016

Corrigendum to “Particle Radiation-Induced Nontargeted Effects in Bone-Marrow-Derived Endothelial Progenitor Cells”

Sharath P. Sasi; Daniel Park; Sujatha Muralidharan; Justin Wage; Albert Kiladjian; Jillian Onufrak; Heiko Enderling; Xinhua Yan; David A. Goukassian

[This corrects the article DOI: 10.1155/2015/496512.].


PLOS ONE | 2015

Correction: Cardiovascular Risks Associated with Low Dose Ionizing Particle Radiation

Xinhua Yan; Sharath P. Sasi; Hannah Gee; Juyong Lee; Yongyao Yang; Raman Mehrzad; Jillian Onufrak; Jin Song; Heiko Enderling; Akhil Agarwal; Layla Rahimi; James P. Morgan; Paul F. Wilson; Joseph P. Carrozza; Kenneth Walsh; Raj Kishore; David A. Goukassian

In the Funding section, the grant number from the funder National Aeronautic and Space Administration has been updated by the funder. The updated grant number is: NNX11AD22G.


Molecular Cancer Therapeutics | 2015

Abstract A56: A dual PI3K-mTOR inhibitor induced cardiac hypertrophy in mice: Role of the insulin signaling

Jillian Onufrak; Yongayo Yang; Juyong Lee; John W. Fuseler; Maoyun Sun; Sharath P. Sasi; J. Tyson McDonald; Lynn Hlatky; Robert L. Price; Joseph P. Carrozza; David A. Goukassian; Thomas K. Borg; James P. Morgan; Xinhua Yan

Studies from us and others showed that mice with cardiomyocyte-specific overexpression of a dominant negative PI3Kp110α had smaller hearts but maintained normal cardiac function. Yet, these mice developed systolic heart failure in the presence of cardiac stress, such as chemotherapy drug doxorubicin and aortic stenosis. Currently, drugs targeting the PI3K pathway are being tested in clinical trials for cancer therapy. Therefore, it is important to assess the cardiac effects of pharmacological inhibition of this pathway. Here, we report that a dual PI3K-mTOR inhibitor, BEZ235 (BEZ), induced cardiac hypertrophy and subsequent heart failure in mice when it was used alone or in combination with doxorubicin. This effect of BEZ was associated with increased hepatic gluconeogenesis, hyperglycemia, hyperinsulinemia and increased activation of the IGFR/insulin receptor (IR) in the heart. In isolated cardiomyocyte culture, BEZ induced feedback activation of IGFR/IR, but this effect was negligible compared to the IGFR/IR activation in the presence of insulin. Injections of insulin in mice lowered blood glucose, improved glucose and pyruvate tolerance but further increased the activation of IGFR/IR in the heart and aggravated BEZ-induced cardiac dysfunction; whereas OSI-906, an IGFR/IR inhibitor, alleviated this cardiac dysfunction. In addition, low dose Metformin significantly prolonged survival and improved cardiac function in BEZ-treated mice. Taken together, these results identified hyperinsulinemia, a systematic effect of the drug, as one of the mechanisms of BEZ-induced cardiac hypertrophy, arguing animal studies are indispensable for evaluating drug toxicity. They also suggest that Metformin but not insulin should be considered for controlling hyperglycemia and preventing the cardiac side effects that may be caused by PI3K pathway inhibitors in clinic. Citation Format: Jillian Onufrak, Yongayo Yang, Juyong Lee, John Fuseler, Maoyun Sun, Sharath Sasi, J. Tyson McDonald, Lynn Hlatky, Robert L. Price, Joseph Carrozza, David Goukassian, Thomas K. Borg, James P. Morgan, Xinhua Yan. A dual PI3K-mTOR inhibitor induced cardiac hypertrophy in mice: Role of the insulin signaling. [abstract]. In: Proceedings of the AACR Special Conference: Targeting the PI3K-mTOR Network in Cancer; Sep 14-17, 2014; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(7 Suppl):Abstract nr A56.


Journal of Cardiovascular Diseases and Diagnosis | 2013

Resistin Is a Novel Biomarker for a Risk of Heart Failure

Yun Bian; Marcy Silver; Jillian Onufrak; Kalon K.L. Ho; Mark A. Marchionni; Peter M. Kang; David A. Goukassian; Joseph P. Carrozza; James P. Morgan; Xinhua Yan

Resistin is derived mainly from fat tissue in rodents, and serum levels are elevated in animal models of obesity and insulin resistance. Recent clinical studies have demonstrated that circulating resistin is associated with inflammation, coronary atherosclerosis, renal dysfunction, adverse prognosis in patients with atherothrombotic ischemic stroke and heart failure. In the population-based observational study, increased circulating levels of resistin are associated with incidence of new-onset heart failure, even after accounting for prevalent coronary heart disease, obesity, insulin resistance, and inflammation. In the elderly, incident heart failure rates (per 1000 person-years) elevate with increasing baseline resistin concentrations. In addition, resistin is strongly associated with risk for incident heart failure in Cox proportional hazard models controlling for clinical variables, inflammatory biomarkers, and measures of adiposity.


Circulation Research | 2014

Abstract 71: A Dual PI3K-mTOR Inhibitor Induced Hyperglycemia and Increased Insulin Signals in the Heart

Xinhua Yan; Jillian Onufrak; Yongyao Yang; Juyong Lee; John W. Fuseler; Maoyun Sun; Sharath P. Sasi; David A. Goukassian; Lynn Hlatky; Robert L. Price; Joseph P. Carrozza; Thomas K. Borg; James P. Morgan

Collaboration


Dive into the Jillian Onufrak's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James P. Morgan

Prince Henry's Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Juyong Lee

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John W. Fuseler

University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Robert L. Price

University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge