Network


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

Hotspot


Dive into the research topics where Danielle Twaroski is active.

Publication


Featured researches published by Danielle Twaroski.


Anesthesiology | 2014

Down-regulation of microRNA-21 is involved in the propofol-induced neurotoxicity observed in human stem cell-derived neurons.

Danielle Twaroski; Yasheng Yan; Jessica Olson; Zeljko J. Bosnjak; Xiaowen Bai

Background:Recent studies in various animal models have suggested that anesthetics such as propofol, when administered early in life, can lead to neurotoxicity. These studies have raised significant safety concerns regarding the use of anesthetics in the pediatric population and highlight the need for a better model to study anesthetic-induced neurotoxicity in humans. Human embryonic stem cells are capable of differentiating into any cell type and represent a promising model to study mechanisms governing anesthetic-induced neurotoxicity. Methods:Cell death in human embryonic stem cell–derived neurons was assessed using terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate in situ nick end labeling staining, and microRNA expression was assessed using quantitative reverse transcription polymerase chain reaction. miR-21 was overexpressed and knocked down using an miR-21 mimic and antagomir, respectively. Sprouty 2 was knocked down using a small interfering RNA, and the expression of the miR-21 targets of interest was assessed by Western blot. Results:Propofol dose and exposure time dependently induced significant cell death (n = 3) in the neurons and down-regulated several microRNAs, including miR-21. Overexpression of miR-21 and knockdown of Sprouty 2 attenuated the increase in terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate in situ nick end labeling–positive cells following propofol exposure. In addition, miR-21 knockdown increased the number of terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate in situ nick end labeling–positive cells by 30% (n = 5). Finally, activated signal transducer and activator of transcription 3 and protein kinase B (Akt) were down-regulated, and Sprouty 2 was up-regulated following propofol exposure (n = 3). Conclusions:These data suggest that (1) human embryonic stem cell–derived neurons represent a promising in vitro human model for studying anesthetic-induced neurotoxicity, (2) propofol induces cell death in human embryonic stem cell–derived neurons, and (3) the propofol-induced cell death may occur via a signal transducer and activator of transcription 3/miR-21/Sprouty 2–dependent mechanism.


Anesthesiology | 2015

Altered Mitochondrial Dynamics Contributes to Propofol-induced Cell Death in Human Stem Cell–derived Neurons

Danielle Twaroski; Yasheng Yan; Ivan Zaja; Eric Clark; Zeljko J. Bosnjak; Xiaowen Bai

Background: Studies in developing animals have shown that anesthetic agents can lead to neuronal cell death and learning disabilities when administered early in life. Development of human embryonic stem cell–derived neurons has provided a valuable tool for understanding the effects of anesthetics on developing human neurons. Unbalanced mitochondrial fusion and fission lead to various pathological conditions including neurodegeneration. The aim of this study was to dissect the role of mitochondrial dynamics in propofol-induced neurotoxicity. Methods: Terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate in situ nick-end labeling staining was used to assess cell death in human embryonic stem cell–derived neurons. Mitochondrial fission was assessed using TOM20 staining and electron microscopy. Expression of mitochondrial fission-related proteins was assessed by Western blot, and confocal microscopy was used to assess opening time of the mitochondrial permeability transition pore (mPTP). Results: Exposure to 6 h of 20 &mgr;g/ml propofol increased cell death from 3.18 ± 0.17% in the control-treated group to 9.6 ± 0.95% and led to detrimental increases in mitochondrial fission (n = 5 coverslips per group) accompanied by increased expression of activated dynamin-related protein 1 and cyclin-dependent kinase 1, key proteins responsible for mitochondrial fission. Propofol exposure also induced earlier opening of the mPTP from 118.9 ± 3.1 s in the control-treated group to 73.3 ± 1.6 s. Pretreatment of the cells with mdivi-1, a mitochondrial fission blocker rescued the propofol-induced toxicity, mitochondrial fission, and mPTP opening time (n = 75 cells per group). Inhibiting cyclin-dependent kinase 1 attenuated the increase in cell death and fission and the increase in expression of activated dynamin-related protein 1. Conclusion: These data demonstrate for the first time that propofol-induced neurotoxicity occurs through a mitochondrial fission/mPTP-mediated pathway.


Seminars in Cardiothoracic and Vascular Anesthesia | 2013

Modeling anesthetic developmental neurotoxicity using human stem cells.

Xiaowen Bai; Danielle Twaroski; Zeljko J. Bosnjak

Mounting preclinical evidence in rodents and nonhuman primates has demonstrated that prolonged exposure of developing animals to general anesthetics can induce widespread neuronal cell death followed by long-term memory and learning disabilities. In vitro experimental evidence from cultured neonatal animal neurons confirmed the in vivo findings. However, there is no direct clinical evidence of the detrimental effects of anesthetics in human fetuses, infants, or children. Development of an in vitro neurogenesis system using human stem cells has opened up avenues of research for advancing our understanding of human brain development and the issues relevant to anesthetic-induced developmental toxicity in human neuronal lineages. Recent studies from our group, as well as other groups, showed that isoflurane influences human neural stem cell proliferation and neurogenesis, whereas ketamine induces neuroapoptosis. Application of this high throughput in vitro stem cell neurogenesis approach is a major stride toward ensuring the safety of anesthetic agents in young children. This in vitro human model allows us to (1) screen the toxic effects of various anesthetics under controlled conditions during intense neuronal growth, (2) find the trigger for the anesthetic-induced catastrophic chain of toxic events, and (3) develop prevention strategies to avoid this toxic effect. In this article, we reviewed the current findings in anesthetic-induced neurotoxicity studies, specifically focusing on the in vitro human stem cell model.


Anesthesiology | 2015

Up-regulation of MicroRNA-21 Mediates Isoflurane-induced Protection of Cardiomyocytes

Jessica Olson; Yasheng Yan; Xiaowen Bai; Zhi-Dong Ge; Mingyu Liang; Alison J. Kriegel; Danielle Twaroski; Zeljko J. Bosnjak

Background:Anesthetic cardioprotection reduces myocardial infarct size after ischemia–reperfusion injury. Currently, the role of microRNA in this process remains unknown. MicroRNAs are short, noncoding nucleotide sequences that negatively regulate gene expression through degradation or suppression of messenger RNA. In this study, the authors uncovered the functional role of microRNA-21 (miR-21) up-regulation after anesthetic exposure. Methods:MicroRNA and messenger RNA expression changes were analyzed by quantitative real-time polymerase chain reaction in cardiomyocytes after exposure to isoflurane. Lactate dehydrogenase release assay and propidium iodide staining were conducted after inhibition of miR-21. miR-21 target expression was analyzed by Western blot. The functional role of miR-21 was confirmed in vivo in both wild-type and miR-21 knockout mice. Results:Isoflurane induces an acute up-regulation of miR-21 in both in vivo and in vitro rat models (n = 6, 247.8 ± 27.5% and 258.5 ± 9.0%), which mediates protection to cardiomyocytes through down-regulation of programmed cell death protein 4 messenger RNA (n = 3, 82.0 ± 4.9% of control group). This protective effect was confirmed by knockdown of miR-21 and programmed cell death protein 4 in vitro. In addition, the protective effect of isoflurane was abolished in miR-21 knockout mice in vivo, with no significant decrease in infarct size compared with nonexposed controls (n = 8, 62.3 ± 4.6% and 56.2 ± 3.2%). Conclusions:The authors demonstrate for the first time that isoflurane mediates protection of cardiomyocytes against oxidative stress via an miR-21/programmed cell death protein 4 pathway. These results reveal a novel mechanism by which the damage done by ischemia/reperfusion injury may be decreased.


Anesthesia & Analgesia | 2016

High Glucose Attenuates Anesthetic Cardioprotection in Stem-Cell-Derived Cardiomyocytes: The Role of Reactive Oxygen Species and Mitochondrial Fission.

Scott Canfield; Ivan Zaja; Brian Godshaw; Danielle Twaroski; Xiaowen Bai; Zeljko J. Bosnjak

BACKGROUND:Hyperglycemia can blunt the cardioprotective effects of isoflurane in the setting of ischemia–reperfusion injury. Previous studies suggest that reactive oxygen species (ROS) and increased mitochondrial fission play a role in cardiomyocyte death during ischemia–reperfusion injury. To investigate the role of glucose concentration in ROS production and mitochondrial fission during ischemia–reperfusion (with and without anesthetic protection), we used the novel platform of human-induced pluripotent stem-cell (iPSC)–derived cardiomyocytes (CMs). METHODS:Cardiomyocyte differentiation from iPSC was characterized by the expression of CM-specific markers using immunohistochemistry and by measuring contractility. iPSC-CMs were exposed to varying glucose conditions (5, 11, and 25 mM) for 24 hours. Mitochondrial permeability transition pore opening, cell viability, and ROS generation endpoints were used to assess the effects of various treatment conditions. Mitochondrial fission was monitored by the visualization of fragmented mitochondria using confocal microscopy. Expression of activated dynamin-related protein 1, a key protein responsible for mitochondrial fission, was assessed by Western blot. RESULTS:Cardiomyocytes were successfully differentiated from iPSC. Elevated glucose conditions (11 and 25 mM) significantly increased ROS generation, whereas only the 25-mM high glucose condition induced mitochondrial fission and increased the expression of activated dynamin-related protein 1 in iPSC-CMs. Isoflurane delayed mitochondrial permeability transition pore opening and protected iPSC-CMs from oxidative stress in 5- and 11-mM glucose conditions to a similar level as previously observed in various isolated animal cardiomyocytes. Scavenging ROS with Trolox or inhibiting mitochondrial fission with mdivi-1 restored the anesthetic cardioprotective effects in iPSC-CMs in 25-mM glucose conditions. CONCLUSIONS:Human iPSC-CM is a useful, relevant model for studying isoflurane cardioprotection and can be manipulated to recapitulate complex clinical perturbations. We demonstrate that the cardioprotective effects of isoflurane in elevated glucose conditions can be restored by scavenging ROS or inhibiting mitochondrial fission. These findings may contribute to further understanding and guidance for restoring pharmacological cardioprotection in hyperglycemic patients.


Pharmaceutica Analytica Acta | 2015

MicroRNAs: New Players in Anesthetic-Induced Developmental Neurotoxicity

Danielle Twaroski; Zeljko J. Bosnjak; Xiaowen Bai

Growing evidence demonstrates that prolonged exposure to general anesthetics during brain development induces widespread neuronal cell death followed by long-term memory and learning disabilities in animal models. These studies have raised serious concerns about the safety of anesthetic use in pregnant women and young children. However, the underlying mechanisms of anesthetic-induced neurotoxicity are complex and are not well understood. MicroRNAs are endogenous, small, non-coding RNAs that have been implicated to play important roles in many different disease processes by negatively regulating target gene expression. A possible role for microRNAs in anesthetic-induced developmental neurotoxicity has recently been identified, suggesting that microRNA-based signaling might be a novel target for preventing the neurotoxicity. Here we provide an overview of anesthetic-induced developmental neurotoxicity and focus on the role of microRNAs in the neurotoxicity observed in both human stem cell-derived neuron and animal models. Aberrant expression of some microRNAs has been shown to be involved in anesthetic-induced developmental neurotoxicity, revealing the potential of microRNAs as therapeutic or preventive targets against the toxicity.


Archive | 2018

Ketamine Induces Neuroapoptosis in Stem Cell–Derived Developing Human Neurons Possibly through Intracellular Calcium/Mitochondria/microRNA Signaling Pathway

Danielle Twaroski; Yasheng Yan; Congshan Jiang; Sarah Logan; Zeljko J. Bosnjak; Xiaowen Bai


Archive | 2016

Analysis of MicroRNAs and their Potential Targets in Human Embryonic Stem Cell-Derived Neurons Treated with the Anesthetic Propofol

Danielle Twaroski; Yasheng Yan; Jessica Olson; Mingyu Liang; Zeljko J. Bosnjak; Xiaowen Bai


The FASEB Journal | 2014

The role of miR-21 in propofol-induced neurotoxicity in developing human neurons (1093.2)

Danielle Twaroski; Xiaowen Bai; Jessica Olson; Yasheng Yan; Yanan Liu; Zeljko J. Bosnjak


The FASEB Journal | 2014

The role of microRNA-21 in anesthetic-mediated cardioprotection (1155.9)

Jessica Olson; Yasheng Yan; Danielle Twaroski; Scott Canfield; Chika Kikuchi; Alison J. Kriegel; Xiaowen Bai; Mingyu Liang; Zeljko J. Bosnjak

Collaboration


Dive into the Danielle Twaroski's collaboration.

Top Co-Authors

Avatar

Xiaowen Bai

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Zeljko J. Bosnjak

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Yasheng Yan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jessica Olson

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Mingyu Liang

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Scott Canfield

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Alison J. Kriegel

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Chika Kikuchi

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Ivan Zaja

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Zhi-Dong Ge

Medical College of Wisconsin

View shared research outputs
Researchain Logo
Decentralizing Knowledge