32 results on '"Molinar-Inglis, Olivia"'
Search Results
2. Phosphoproteomic analysis of thrombin- and p38 MAPK-regulated signaling networks in endothelial cells
- Author
-
Molinar-Inglis, Olivia, Wozniak, Jacob M, Grimsey, Neil J, Orduña-Castillo, Lennis B, Cheng, Norton, Lin, Ying, Ramirez, Monica L Gonzalez, Birch, Cierra A, Lapek, John D, Gonzalez, David J, and Trejo, JoAnn
- Subjects
Biochemistry and Cell Biology ,Medical Physiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Cancer ,Hematology ,2.1 Biological and endogenous factors ,Aetiology ,1.1 Normal biological development and functioning ,Underpinning research ,Cells ,Cultured ,Endothelial Cells ,Humans ,MAP Kinase Signaling System ,Phosphorylation ,Proteomics ,Receptor ,PAR-1 ,Thrombin ,p38 Mitogen-Activated Protein Kinases ,ERK1/2 ,GPCR ,inflammation ,protein kinase ,α-catenin ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
Endothelial dysfunction is a hallmark of inflammation and is mediated by inflammatory factors that signal through G protein-coupled receptors including protease-activated receptor-1 (PAR1). PAR1, a receptor for thrombin, signals via the small GTPase RhoA and myosin light chain intermediates to facilitate endothelial barrier permeability. PAR1 also induces endothelial barrier disruption through a p38 mitogen-activated protein kinase-dependent pathway, which does not integrate into the RhoA/MLC pathway; however, the PAR1-p38 signaling pathways that promote endothelial dysfunction remain poorly defined. To identify effectors of this pathway, we performed a global phosphoproteome analysis of thrombin signaling regulated by p38 in human cultured endothelial cells using multiplexed quantitative mass spectrometry. We identified 5491 unique phosphopeptides and 2317 phosphoproteins, four distinct dynamic phosphoproteome profiles of thrombin-p38 signaling, and an enrichment of biological functions associated with endothelial dysfunction, including modulators of endothelial barrier disruption and a subset of kinases predicted to regulate p38-dependent thrombin signaling. Using available antibodies to detect identified phosphosites of key p38-regulated proteins, we discovered that inhibition of p38 activity and siRNA-targeted depletion of the p38α isoform increased basal phosphorylation of extracellular signal-regulated protein kinase 1/2, resulting in amplified thrombin-stimulated extracellular signal-regulated protein kinase 1/2 phosphorylation that was dependent on PAR1. We also discovered a role for p38 in the phosphorylation of α-catenin, a component of adherens junctions, suggesting that this phosphorylation may function as an important regulatory process. Taken together, these studies define a rich array of thrombin- and p38-regulated candidate proteins that may serve important roles in endothelial dysfunction.
- Published
- 2022
3. aPC/PAR1 confers endothelial anti-apoptotic activity via a discrete, β-arrestin-2–mediated SphK1-S1PR1-Akt signaling axis
- Author
-
Molinar-Inglis, Olivia, Birch, Cierra A, Nicholas, Dequina, Orduña-Castillo, Lennis, Cisneros-Aguirre, Metztli, Patwardhan, Anand, Chen, Buxin, Grimsey, Neil J, Coronel, Luisa J, Lin, Huilan, Menzies, Patrick K Gomez, Lawson, Mark A, Patel, Hemal H, and Trejo, JoAnn
- Subjects
Anilides ,Apoptosis ,Endothelial Cells ,Enzyme Inhibitors ,Gene Expression Regulation ,Heterocyclic Compounds ,3-Ring ,Humans ,Lactones ,Methanol ,Organophosphonates ,Phosphotransferases (Alcohol Group Acceptor) ,Platelet Aggregation Inhibitors ,Protein C ,Proto-Oncogene Proteins c-akt ,Pyridines ,Pyrrolidines ,Receptor ,PAR-1 ,Sphingosine-1-Phosphate Receptors ,Sulfones ,beta-Arrestin 2 ,biased signaling ,cytoprotection ,GPCR ,endothelial dysfunction - Abstract
Endothelial dysfunction is associated with vascular disease and results in disruption of endothelial barrier function and increased sensitivity to apoptosis. Currently, there are limited treatments for improving endothelial dysfunction. Activated protein C (aPC), a promising therapeutic, signals via protease-activated receptor-1 (PAR1) and mediates several cytoprotective responses, including endothelial barrier stabilization and anti-apoptotic responses. We showed that aPC-activated PAR1 signals preferentially via β-arrestin-2 (β-arr2) and dishevelled-2 (Dvl2) scaffolds rather than G proteins to promote Rac1 activation and barrier protection. However, the signaling pathways utilized by aPC/PAR1 to mediate anti-apoptotic activities are not known. aPC/PAR1 cytoprotective responses also require coreceptors; however, it is not clear how coreceptors impact different aPC/PAR1 signaling pathways to drive distinct cytoprotective responses. Here, we define a β-arr2-mediated sphingosine kinase-1 (SphK1)-sphingosine-1-phosphate receptor-1 (S1PR1)-Akt signaling axis that confers aPC/PAR1-mediated protection against cell death. Using human cultured endothelial cells, we found that endogenous PAR1 and S1PR1 coexist in caveolin-1 (Cav1)-rich microdomains and that S1PR1 coassociation with Cav1 is increased by aPC activation of PAR1. Our study further shows that aPC stimulates β-arr2-dependent SphK1 activation independent of Dvl2 and is required for transactivation of S1PR1-Akt signaling and protection against cell death. While aPC/PAR1-induced, extracellular signal-regulated kinase 1/2 (ERK1/2) activation is also dependent on β-arr2, neither SphK1 nor S1PR1 are integrated into the ERK1/2 pathway. Finally, aPC activation of PAR1-β-arr2-mediated protection against apoptosis is dependent on Cav1, the principal structural protein of endothelial caveolae. These studies reveal that different aPC/PAR1 cytoprotective responses are mediated by discrete, β-arr2-driven signaling pathways in caveolae.
- Published
- 2021
4. Subcellular hot spots of GPCR signaling promote vascular inflammation
- Author
-
Birch, Cierra A, Molinar-Inglis, Olivia, and Trejo, JoAnn
- Subjects
Medical Biochemistry and Metabolomics ,Biomedical and Clinical Sciences ,Clinical Sciences ,2.1 Biological and endogenous factors ,Arrestins ,B-cell lymphoma protein 10 ,COVID-19 ,Endosomes ,Endothelial ,G protein-coupled receptor ,GPCR ,JAK-STAT ,Janus kinase ,JAK ,MALT1 ,NFκB ,adherens junctions ,AJ ,angiotensin II type 1 receptor ,AT1 ,angiotensin converting enzyme-2 ,ACE2 ,caspase recruitment domain-containing protein ,CARMA ,coronavirus disease of 2019 ,COVID-19 ,fibroblast-growth-factor ,FGF ,inhibitor of NFκB kinase ,IKK ,mitogen-activated protein kinase ,MAPK ,mucosa-associated lymphoid tissue lymphoma translocation protein 1 ,neural precursor cell expressed developmentally downregulated protein 4 ,NEDD4 ,nuclear factor kappa-light-chain-enhancer of activated B cells ,NFκB ,p38 MAPK ,platelet activating factor ,PAF ,protease-activated receptor-1 ,PAR1 ,severe acute respiratory syndrome coronavirus 2 ,SARS-CoV-2 ,signal transducer and activator of transcription ,STAT ,transforming growth factor-α-activated kinase binding protein-1 ,TAB1 ,Clinical sciences ,Medical biochemistry and metabolomics - Abstract
G-coupled protein receptors (GPCRs) comprise the largest class of druggable targets. Signaling by GPCRs is initiated from subcellular hot spots including the plasma membrane, signalosomes, and endosomes to contribute to vascular inflammation. GPCR-G protein signaling at the plasma membrane causes endothelial barrier disruption and also cross-talks with growth factor receptors to promote proinflammatory signaling. A second surge of GPCR signaling is initiated by cytoplasmic NFκB activation mediated by β-arrestins and CARMA-BCL10-MALT1 signalosomes. Once internalized, ubiquitinated GPCRs initiate signaling from endosomes via assembly of the transforming growth factor-β-activated kinase binding protein-1 (TAB1)-TAB2-p38 MAPK complex to promote vascular inflammation. Understanding the complexities of GPCR signaling is critical for development of new strategies to treat vascular inflammation such as that associated with COVID-19.
- Published
- 2021
5. Location-specific inhibition of Akt reveals regulation of mTORC1 activity in the nucleus.
- Author
-
Zhou, Xin, Zhong, Yanghao, Molinar-Inglis, Olivia, Kunkel, Maya T, Chen, Mingyuan, Sun, Tengqian, Zhang, Jiao, Shyy, John Y-J, Trejo, JoAnn, Newton, Alexandra C, and Zhang, Jin
- Subjects
NIH 3T3 Cells ,Cell Nucleus ,Animals ,Humans ,Mice ,Phosphoproteins ,Protein Kinase Inhibitors ,Signal Transduction ,Proto-Oncogene Proteins c-akt ,Gene Knockdown Techniques ,HEK293 Cells ,TOR Serine-Threonine Kinases ,Mechanistic Target of Rapamycin Complex 1 ,1.1 Normal biological development and functioning ,Generic health relevance - Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) integrates growth, nutrient and energy status cues to control cell growth and metabolism. While mTORC1 activation at the lysosome is well characterized, it is not clear how this complex is regulated at other subcellular locations. Here, we combine location-selective kinase inhibition, live-cell imaging and biochemical assays to probe the regulation of growth factor-induced mTORC1 activity in the nucleus. Using a nuclear targeted Akt Substrate-based Tandem Occupancy Peptide Sponge (Akt-STOPS) that we developed for specific inhibition of Akt, a critical upstream kinase, we show that growth factor-stimulated nuclear mTORC1 activity requires nuclear Akt activity. Further mechanistic dissection suggests that nuclear Akt activity mediates growth factor-induced nuclear translocation of Raptor, a regulatory scaffolding component in mTORC1, and localization of Raptor to the nucleus results in nuclear mTORC1 activity in the absence of growth factor stimulation. Taken together, these results reveal a mode of regulation of mTORC1 that is distinct from its lysosomal activation, which controls mTORC1 activity in the nuclear compartment.
- Published
- 2020
6. GLUT1-mediated glycolysis supports GnRH-induced secretion of luteinizing hormone from female gonadotropes.
- Author
-
Nicholas, Dequina A, Knight, Vashti S, Tonsfeldt, Karen J, Terasaka, Tomohiro, Molinar-Inglis, Olivia, Stephens, Shannon BZ, Trejo, JoAnn, Kauffman, Alexander S, Mellon, Pamela L, and Lawson, Mark A
- Subjects
Cells ,Cultured ,Animals ,Mice ,Inbred C57BL ,Luteinizing Hormone ,Glucose ,Receptors ,LHRH ,RNA ,Messenger ,Glycolysis ,Female ,Glucose Transporter Type 1 ,Gonadotropin-Releasing Hormone ,Gonadotrophs ,Neurosciences ,Contraception/Reproduction ,1.1 Normal biological development and functioning ,Reproductive health and childbirth - Abstract
The mechanisms mediating suppression of reproduction in response to decreased nutrient availability remain undefined, with studies suggesting regulation occurs within the hypothalamus, pituitary, or gonads. By manipulating glucose utilization and GLUT1 expression in a pituitary gonadotrope cell model and in primary gonadotropes, we show GLUT1-dependent stimulation of glycolysis, but not mitochondrial respiration, by the reproductive neuropeptide GnRH. GnRH stimulation increases gonadotrope GLUT1 expression and translocation to the extracellular membrane. Maximal secretion of the gonadotropin Luteinizing Hormone is supported by GLUT1 expression and activity, and GnRH-induced glycolysis is recapitulated in primary gonadotropes. GLUT1 expression increases in vivo during the GnRH-induced ovulatory LH surge and correlates with GnRHR. We conclude that the gonadotropes of the anterior pituitary sense glucose availability and integrate this status with input from the hypothalamus via GnRH receptor signaling to regulate reproductive hormone synthesis and secretion.
- Published
- 2020
7. Signaling diversity enabled by Rap1-regulated plasma membrane ERK with distinct temporal dynamics.
- Author
-
Keyes, Jeremiah, Ganesan, Ambhighainath, Molinar-Inglis, Olivia, Hamidzadeh, Archer, Zhang, Jinfan, Ling, Megan, Trejo, JoAnn, Levchenko, Andre, and Zhang, Jin
- Subjects
PC12 Cells ,Animals ,Humans ,Rats ,Epidermal Growth Factor ,rap1 GTP-Binding Proteins ,rac1 GTP-Binding Protein ,Extracellular Signal-Regulated MAP Kinases ,HEK293 Cells ,EGF ,ERK ,PC12 ,Rap1 ,biochemistry ,cell biology ,chemical biology ,none ,signal transduction ,spatiotemporal ,Bioengineering ,1.1 Normal biological development and functioning ,Biochemistry and Cell Biology - Abstract
A variety of different signals induce specific responses through a common, extracellular-signal regulated kinase (ERK)-dependent cascade. It has been suggested that signaling specificity can be achieved through precise temporal regulation of ERK activity. Given the wide distrubtion of ERK susbtrates across different subcellular compartments, it is important to understand how ERK activity is temporally regulated at specific subcellular locations. To address this question, we have expanded the toolbox of Förster Resonance Energy Transfer (FRET)-based ERK biosensors by creating a series of improved biosensors targeted to various subcellular regions via sequence specific motifs to measure spatiotemporal changes in ERK activity. Using these sensors, we showed that EGF induces sustained ERK activity near the plasma membrane in sharp contrast to the transient activity observed in the cytoplasm and nucleus. Furthermore, EGF-induced plasma membrane ERK activity involves Rap1, a noncanonical activator, and controls cell morphology and EGF-induced membrane protrusion dynamics. Our work strongly supports that spatial and temporal regulation of ERK activity is integrated to control signaling specificity from a single extracellular signal to multiple cellular processes.
- Published
- 2020
8. MON-009 GLUT1-Mediated Glycolysis Facilitates GnRH-Induced Secretion of Luteinizing Hormone
- Author
-
Nicholas, Dequina, Knight, Vashti, Tonsfeldt, Karen, Terasaka, Tomohiro, Molinar-Inglis, Olivia, Stephens, Shannon Brooke Zoe, Trejo, Joann, Kauffman, Alexander S, Mellon, Pamela L, and Lawson, Mark A
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Neurosciences ,Contraception/Reproduction ,Diabetes ,1.1 Normal biological development and functioning ,Underpinning research ,Cardiovascular medicine and haematology - Abstract
Abstract: Reproduction requires intensive energy expenditure, and energy availability impacts the function of the reproductive endocrine HPG-axis. Accordingly, the reproductive axis is suppressed during hypoglycemia. Circulating blood glucose can directly interact with gonadotropes within the highly vascular pituitary. Therefore, it is possible that gonadotropes may sense energy availability via the presence of glucose in the circulation and integrate this status with input from GnRH neurons to regulate hormone production. Gonadotropes dominantly express glucose transporter 1 (GLUT1) and increase glucose uptake in response to GnRH. Thus, we hypothesized that gonadotropes engage glycolysis in response to GnRH stimulation due to the high energy demand of protein synthesis required for LH production. We developed an approach to sort and successfully culture primary gonadotropes from wild type mice. Using this approach, we performed extracellular flux analysis and found that gonadotropes respond to GnRH by inducing GLUT1-mediated glycolysis that is independent of mitochondrial respiration. Knock-down of GLUT1 expression in the LβT2 gonadotrope cell line, glucose restriction, or treatment with the competitive inhibitor of glycolysis, 2-DG, diminished GnRH-induced LH secretion, indicating GLUT1 expression is necessary for maximal GnRH-induced LH secretion. We confirmed this observation in primary female mouse gonadotropes by limiting glucose availability which resulted in diminished basal LH and FSH secretion. Lastly, GLUT1 expression in the pituitary correlates with GnRH receptor expression and is increased during the LH surge in a mouse model. These results implicate glucose uptake through GLUT1 as permissive for gonadotrope secretion of LH and therefore reproductive function, especially the LH surge. We conclude that GLUT1-mediated glucose uptake is an important rate-limiting step in gonadotropin synthesis and operation of the HPG-axis.
- Published
- 2020
9. Phosphoproteomic analysis of protease-activated receptor-1 biased signaling reveals unique modulators of endothelial barrier function
- Author
-
Lin, Ying, Wozniak, Jacob M, Grimsey, Neil J, Girada, Sravan, Patwardhan, Anand, Molinar-Inglis, Olivia, Smith, Thomas H, Lapek, John D, Gonzalez, David J, and Trejo, JoAnn
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Hematology ,HIV/AIDS ,2.1 Biological and endogenous factors ,Aetiology ,Calmodulin-Binding Proteins ,Carrier Proteins ,Endothelial Cells ,Humans ,Microfilament Proteins ,Phosphorylation ,Protein C Inhibitor ,Proteomics ,Receptor ,PAR-1 ,Signal Transduction ,Thrombin ,actin ,arrestin ,GPCR ,inflammation ,thrombin - Abstract
Thrombin, a procoagulant protease, cleaves and activates protease-activated receptor-1 (PAR1) to promote inflammatory responses and endothelial dysfunction. In contrast, activated protein C (APC), an anticoagulant protease, activates PAR1 through a distinct cleavage site and promotes anti-inflammatory responses, prosurvival, and endothelial barrier stabilization. The distinct tethered ligands formed through cleavage of PAR1 by thrombin versus APC result in unique active receptor conformations that bias PAR1 signaling. Despite progress in understanding PAR1 biased signaling, the proteins and pathways utilized by thrombin versus APC signaling to induce opposing cellular functions are largely unknown. Here, we report the global phosphoproteome induced by thrombin and APC signaling in endothelial cells with the quantification of 11,266 unique phosphopeptides using multiplexed quantitative mass spectrometry. Our results reveal unique dynamic phosphoproteome profiles of thrombin and APC signaling, an enrichment of associated biological functions, including key modulators of endothelial barrier function, regulators of gene transcription, and specific kinases predicted to mediate PAR1 biased signaling. Using small interfering RNA to deplete a subset of phosphorylated proteins not previously linked to thrombin or APC signaling, a function for afadin and adducin-1 actin binding proteins in thrombin-induced endothelial barrier disruption is unveiled. Afadin depletion resulted in enhanced thrombin-promoted barrier permeability, whereas adducin-1 depletion completely ablated thrombin-induced barrier disruption without compromising p38 signaling. However, loss of adducin-1 blocked APC-induced Akt signaling. These studies define distinct thrombin and APC dynamic signaling profiles and a rich array of proteins and biological pathways that engender PAR1 biased signaling in endothelial cells.
- Published
- 2020
10. Gastrointestinal Acute Radiation Syndrome: Mechanisms, Models, Markers, and Medical Countermeasures
- Author
-
Winters, Thomas A., primary, Marzella, Libero, additional, Molinar-Inglis, Olivia, additional, Price, Paul W., additional, Han, Nyun Calvin, additional, Cohen, Jonathan E., additional, Wang, Sue-Jane, additional, Fotenos, Anthony F., additional, Sullivan, Julie M., additional, Esker, John, additional, Lapinskas, Paula J., additional, and DiCarlo, Andrea L., additional
- Published
- 2024
- Full Text
- View/download PDF
11. Subcellular hot spots of GPCR signaling promote vascular inflammation
- Author
-
Birch, Cierra A., Molinar-Inglis, Olivia, and Trejo, JoAnn
- Published
- 2021
- Full Text
- View/download PDF
12. Highlighting the NIAID Radiation and Nuclear Countermeasures Program’s Commitment to Training and Diversifying the Radiation Workforce
- Author
-
Molinar-Inglis, Olivia, primary, Price, LeShawndra N., additional, and DiCarlo, Andrea L., additional
- Published
- 2024
- Full Text
- View/download PDF
13. Radiation-induced multi-organ injury
- Author
-
Molinar-Inglis, Olivia, primary, DiCarlo, Andrea L., additional, Lapinskas, Paula J., additional, Rios, Carmen I., additional, Satyamitra, Merriline M., additional, Silverman, Toby A., additional, Winters, Thomas A., additional, and Cassatt, David R., additional
- Published
- 2024
- Full Text
- View/download PDF
14. Product Development within the National Institutes of Health Radiation and Nuclear Countermeasures Program.
- Author
-
Cassatt, David R., DiCarlo, Andrea L., and Molinar-Inglis, Olivia
- Subjects
NEW product development ,MEDICAL research ,RADIATION injuries ,INVESTIGATIONAL drugs ,SKIN injuries - Abstract
The Radiation and Nuclear Countermeasures Program (RNCP) at the National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH) was established to facilitate the development of medical countermeasures (MCMs) and diagnostic approaches for use in a radiation public health emergency. Approvals for MCMs can be very challenging but are made possible under the United States Food and Drug Administration (FDA) Animal Rule, which is designed to enable licensure of drugs or biologics when clinical efficacy studies are unethical or unfeasible. The NIAID portfolio includes grants, contracts, and inter-agency agreements designed to span all aspects of drug development and encompasses basic research through FDA approval. In addition, NIAID manages an active portfolio of biodosimetry approaches to assess injuries and absorbed radiation levels to guide triage and treatment decisions. NIAID, together with grantees, contractors, and other stakeholders with promising products, works to advance candidate MCMs and biodosimetry tools through an established product development pipeline. In addition to managing grants and contracts, NIAID tests promising candidates in our established preclinical animal models, and the NIAID Program Officers work closely with sponsors as product managers to guide them through the process. In addition, a valuable benefit for stakeholders is working with the NIAID Office of Regulatory Affairs, where NIAID coordinates with the FDA to facilitate interactions between sponsors and the agency. Activities funded by NIAID include basic research (e.g., library screens to discover new products, determine early efficacy, and delineate mechanism of action) and the development of small and large animal models of radiation-induced hematopoietic, gastrointestinal, lung, kidney, and skin injury, radiation combined injury, and radionuclide decorporation. NIAID also sponsors Good Laboratory Practice product safety, pharmacokinetic, pharmacodynamic, and toxicology studies, as well as efficacy and dose-ranging studies to optimize product regimens. For later-stage candidates, NIAID funds large-scale manufacturing and formulation development of products. The program also supports Phase 1 human clinical studies to ensure human safety and to bridge pharmacokinetic, pharmacodynamic, and efficacy data from animals to humans. To date, NIAID has supported >900 animal studies and one clinical study, evaluating >500 new/repurposed radiation MCMs and biodosimetric approaches. NIAID sponsorship led to the approval of three of the six drugs for acute radiation syndrome under the FDA Animal Rule, five Investigational New Drug applications, and 18 additional submissions for Investigational Device Exemptions, while advancing 38 projects to the Biomedical Advanced Research and Development Authority for follow-on research and development. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. The NIAID/RNCP Biodosimetry Program: An Overview.
- Author
-
Satyamitra, Merriline M., Cassatt, David R., Molinar-Inglis, Olivia, Rios, Carmen I., Taliaferro, Lanyn P., Winters, Thomas A., and DiCarlo, Andrea L.
- Subjects
RADIATION injuries ,RADIATION exposure ,DOSE-response relationship (Radiation) ,RESEARCH awards ,INNOVATIONS in business ,RESEARCH personnel - Abstract
Established in 2004, the Radiation and Nuclear Countermeasures Program (RNCP), within the National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health has the central mission to advance medical countermeasure mitigators/therapeutics, and biomarkers and technologies to assess, triage, and inform medical management of patients experiencing acute radiation syndrome and/or the delayed effects of acute radiation exposure. The RNCP biodosimetry mission space encompasses: (1) basic research to elucidate novel approaches for rapid and accurate assessment of radiation exposure, (2) studies to support advanced development for US Food and Drug Administration (FDA) clearance of promising triage or treatment devices/approaches, (3) characterization of biomarkers and/or assays to determine degree of tissue or organ dose that can predict outcome of radiation injuries (i.e., organ failure, morbidity, and/or mortality), and (4) outreach efforts to facilitate interactions with researchers developing cutting edge biodosimetry approaches. Thus far, no biodosimetry device has been FDA cleared for use during a radiological/nuclear incident. At NIAID, advancement of radiation biomarkers and biodosimetry approaches is facilitated by a variety of funding mechanisms (grants, contracts, cooperative and interagency agreements, and Small Business Innovation Research awards), with the objective of advancing devices and assays toward clearance, as outlined in the FDA's Radiation Biodosimetry Medical Countermeasure Devices Guidance. The ultimate goal of the RNCP biodosimetry program is to develop and establish accurate and reliable biodosimetry tools that will improve radiation preparedness and ultimately save lives during a radiological or nuclear incident. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. Sex differences in radiation research.
- Author
-
Taliaferro, Lanyn P., Agarwal, Rajeev K., Coleman, C. Norman, DiCarlo, Andrea L., Hofmeyer, Kimberly A., Loelius, Shannon G., Molinar-Inglis, Olivia, Tedesco, Dana C., and Satyamitra, Merriline M.
- Subjects
SCIENTIFIC knowledge ,RADIATION injuries ,RADIATION exposure ,RADIATION ,WOMEN'S health - Abstract
The Sex Differences in Radiation Research workshop addressed the role of sex as a confounder in radiation research and its implication in real-world radiological and nuclear applications. In April 2022, HHS-wide partners from the Radiation and Nuclear Countermeasures Program, the Office of Research on Women's Health National Institutes of Health Office of Women's Health, U.S. Food and Drug Administration, and the Radiological and Nuclear Countermeasures Branch at the Biomedical Advanced Research and Development Authority conducted a workshop to address the scientific implication and knowledge gaps in understanding sex in basic and translational research. The goals of this workshop were to examine sex differences in 1. Radiation animal models and understand how these may affect radiation medical countermeasure development; 2. Biodosimetry and/or biomarkers used to assess acute radiation syndrome, delayed effects of acute radiation exposure, and/or predict major organ morbidities; 3. medical research that lacks representation from both sexes. In addition, regulatory policies that influence inclusion of women in research, and the gaps that exist in drug development and device clearance were discussed. Finally, real-world sex differences in human health scenarios were also considered. This report provides an overview of the two-day workshop, and open discussion among academic investigators, industry researchers, and U.S. government representatives. This meeting highlighted that current study designs lack the power to determine statistical significance based on sex, and much is unknown about the underlying factors that contribute to these differences. Investigators should accommodate both sexes in all stages of research to ensure that the outcome is robust, reproducible, and accurate, and will benefit public health. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Meeting Report Sex Differences in Radiation Research
- Author
-
Taliaferro, Lanyn P., primary, Agarwal, Rajeev K., additional, Coleman, C. Norman, additional, DiCarlo, Andrea L., additional, Hofmeyer, Kimberly A., additional, Loelius, Shannon G., additional, Molinar-Inglis, Olivia, additional, Tedesco, Dana C., additional, and Satyamitra, Merriline M., additional
- Published
- 2023
- Full Text
- View/download PDF
18. aPC/PAR1 confers endothelial anti‐apoptotic activity via a discrete β‐arrestin‐2 mediated SphK1‐S1PR1‐Akt signaling axis
- Author
-
Orduna‐Castillo, Lennis B., primary, Molinar‐Inglis, Olivia, additional, Nicholas, Dequina, additional, Cisneros‐Aguirre, Metztli, additional, Patwardhan, Anand, additional, Grimsey, Neil, additional, Lawson, Mark A., additional, Patel, Hemal, additional, and Trejo, JoAnn, additional
- Published
- 2022
- Full Text
- View/download PDF
19. aPC/PAR1 confers endothelial anti-apoptotic activity via a discrete β-arrestin-2 mediated SphK1-S1PR1-Akt signaling axis
- Author
-
Molinar-Inglis, Olivia, primary, Birch, Cierra A., additional, Nicholas, Dequina, additional, Cisneros-Aguirre, Metzli, additional, Patwardhan, Anand, additional, Chen, Buxin, additional, Grimsey, Neil J., additional, Gomez-Menzies, Patrick K., additional, Lin, Huilan, additional, Coronel, Luisa J., additional, Lawson, Mark A., additional, Patel, Hemal H., additional, and Trejo, JoAnn, additional
- Published
- 2021
- Full Text
- View/download PDF
20. Signaling diversity enabled by Rap1-regulated plasma membrane ERK with distinct temporal dynamics
- Author
-
Keyes, Jeremiah, primary, Ganesan, Ambhighainath, additional, Molinar-Inglis, Olivia, additional, Hamidzadeh, Archer, additional, Zhang, Jinfan, additional, Ling, Megan, additional, Trejo, JoAnn, additional, Levchenko, Andre, additional, and Zhang, Jin, additional
- Published
- 2020
- Full Text
- View/download PDF
21. Author response: Signaling diversity enabled by Rap1-regulated plasma membrane ERK with distinct temporal dynamics
- Author
-
Keyes, Jeremiah, primary, Ganesan, Ambhighainath, additional, Molinar-Inglis, Olivia, additional, Hamidzadeh, Archer, additional, Zhang, Jinfan, additional, Ling, Megan, additional, Trejo, JoAnn, additional, Levchenko, Andre, additional, and Zhang, Jin, additional
- Published
- 2020
- Full Text
- View/download PDF
22. Role of Protease‐activated receptors in Activated protein C‐mediated anti‐inflammatory responses in endothelial cells
- Author
-
Birch, Cierra A., primary, Molinar-Inglis, Olivia, additional, Lin, Ying, additional, and Trejo, JoAnn, additional
- Published
- 2020
- Full Text
- View/download PDF
23. Phospho‐proteomic Analysis of Protease‐activated Receptor‐1 Biased Signaling Reveals Novel Modulators of Endothelial Barrier Function
- Author
-
Lin, Ying, primary, Wozniak, Jacob, additional, Grimsey, Neil, additional, Girada, Sravan, additional, Patwardan, Anand, additional, Molinar-Inglis, Olivia, additional, Smith, Thomas, additional, Lapek, John, additional, Gonzalez, David, additional, and Trejo, JoAnn, additional
- Published
- 2020
- Full Text
- View/download PDF
24. Signaling Diversity Enabled by Rap1 and cAMP/PKA‐Regulated Plasma Membrane ERK with Distinct Temporal Dynamics
- Author
-
Keyes, Jeremiah, primary, Ganesan, Ambhighainath, additional, Molinar-Inglis, Olivia, additional, Hamidzadeh, Archer, additional, Ling, Megan, additional, Trejo, JoAnn, additional, Levchenko, Andre, additional, and Zhang, Jin, additional
- Published
- 2020
- Full Text
- View/download PDF
25. Improved fractionation method demonstrates phosphorylated Akt is localized to the cytoplasm in LβT2 cells
- Author
-
Garcia, Alexis, primary, Molinar-Inglis, Olivia, additional, Nicholas, Dequina, additional, Keyes, Jeremiah, additional, Adame, Jose, additional, Cisneros, Metztli, additional, Lawson, Mark, additional, Zhang, Jin, additional, and Trejo, JoAnn, additional
- Published
- 2020
- Full Text
- View/download PDF
26. Integration of GPCR‐induced endothelial cytoprotection signaling by β‐arrestin‐2
- Author
-
Molinar-Inglis, Olivia, primary, Zhang, Lili, additional, Nicholas, Dequina, additional, Chen, Buxin, additional, Cisneros, Metztli, additional, Grimsey, Neil J., additional, Coronel, Luisa J., additional, Lin, Ying, additional, Rada, Cara, additional, Chi, Neil, additional, Lawson, Mark A., additional, and Trejo, JoAnn, additional
- Published
- 2020
- Full Text
- View/download PDF
27. G‐protein coupled receptor kinases’ (GRKs) role in APC/PAR1‐induced endothelial cells
- Author
-
Ramirez, Monica Lizeth Gonzalez, primary, Lin, Ying, additional, Molinar-Inglis, Olivia, additional, Birch, Cierra, additional, and Trejo, JoAnn, additional
- Published
- 2020
- Full Text
- View/download PDF
28. Signaling Diversity Enabled by Rap1-Regulated Plasma Membrane ERK with Distinct Temporal Dynamics
- Author
-
Keyes, Jeremiah, primary, Ganesan, Ambhighainath, additional, Molinar-Inglis, Olivia, additional, Hamidzadeh, Archer, additional, Ling, Megan, additional, Trejo, JoAnn, additional, Levchenko, Andre, additional, and Zhang, Jin, additional
- Published
- 2019
- Full Text
- View/download PDF
29. Phosphoproteomic analysis of protease-activated receptor-1 biased signaling reveals unique modulators of endothelial barrier function.
- Author
-
Ying Lin, Wozniak, Jacob M., Grimsey, Neil J., Girada, Sravan, Patwardhan, Anand, Molinar-Inglis, Olivia, Smith, Thomas H., Lapek, John D., Gonzalez, David J., and Trejo, JoAnn
- Subjects
SMALL interfering RNA ,CARRIER proteins ,PROTEIN C ,THROMBIN ,PROTEIN microarrays - Abstract
Thrombin, a procoagulant protease, cleaves and activates proteaseactivated receptor-1 (PAR1) to promote inflammatory responses and endothelial dysfunction. In contrast, activated protein C (APC), an anticoagulant protease, activates PAR1 through a distinct cleavage site and promotes anti-inflammatory responses, prosurvival, and endothelial barrier stabilization. The distinct tethered ligands formed through cleavage of PAR1 by thrombin versus APC result in unique active receptor conformations that bias PAR1 signaling. Despite progress in understanding PAR1 biased signaling, the proteins and pathways utilized by thrombin versus APC signaling to induce opposing cellular functions are largely unknown. Here, we report the global phosphoproteome induced by thrombin and APC signaling in endothelial cells with the quantification of 11,266 unique phosphopeptides using multiplexed quantitative mass spectrometry. Our results reveal unique dynamic phosphoproteome profiles of thrombin and APC signaling, an enrichment of associated biological functions, including key modulators of endothelial barrier function, regulators of gene transcription, and specific kinases predicted to mediate PAR1 biased signaling. Using small interfering RNA to deplete a subset of phosphorylated proteins not previously linked to thrombin or APC signaling, a function for afadin and adducin-1 actin binding proteins in thrombin-induced endothelial barrier disruption is unveiled. Afadin depletion resulted in enhanced thrombin-promoted barrier permeability, whereas adducin-1 depletion completely ablated thrombin-induced barrier disruption without compromising p38 signaling. However, loss of adducin-1 blocked APC-induced Akt signaling. These studies define distinct thrombin and APC dynamic signaling profiles and a rich array of proteins and biological pathways that engender PAR1 biased signaling in endothelial cells. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
30. APC2 associates with the actin cortex through a multipart mechanism to regulate cortical actin organization and dynamics in the Drosophila ovary
- Author
-
Molinar‐Inglis, Olivia, primary, Oliver, Stacie L., additional, Rudich, Paige, additional, Kunttas, Ezgi, additional, and McCartney, Brooke M., additional
- Published
- 2018
- Full Text
- View/download PDF
31. An optimized fractionation method reveals insulin-induced membrane surface localization of GLUT1 to increase glycolysis in LβT2 cells.
- Author
-
Molinar-Inglis O, Wiggins K, Varma A, Del Mundo Z, Adame JM, Cozzo A, Muñoz O, Le UV, Trinh D, Garcia AC, Cisneros-Aguirre M, Gonzalez Ramirez ML, Keyes J, Zhang J, Lawson MA, Trejo J, and Nicholas DA
- Abstract
Insulin is an important regulator of whole-body glucose homeostasis. In insulin sensitive tissues such as muscle and adipose, insulin induces the translocation of glucose transporter 4 (GLUT4) to the cell membrane, thereby increasing glucose uptake. However, insulin also signals in tissues that are not generally associated with glucose homeostasis. In the human reproductive endocrine axis, hyperinsulinemia suppresses the secretion of gonadotropins from gonadotrope cells of the anterior pituitary, thereby linking insulin dysregulation to suboptimal reproductive health. In the mouse, gonadotropes express the insulin receptor which has the canonical signaling response of IRS, AKT, and mTOR activation. However, the functional outcomes of insulin action on gonadotropes are unclear. Here, we demonstrate through use of an optimized cell fractionation protocol that insulin stimulation of the LβT2 gonadotropic cell line results in the unexpected translocation of GLUT1 to the plasma membrane. Using our high purity fractionation protocol, we further demonstrate that though Akt signaling in response to insulin is intact, insulin-induced translocation of GLUT1 occurs independently of Akt activation in LβT2 cells., Competing Interests: Declaration of competing interest None., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
32. The NIAID/RNCP Biodosimetry Program: An Overview.
- Author
-
Satyamitra MM, Cassatt DR, Molinar-Inglis O, Rios CI, Taliaferro LP, Winters TA, and DiCarlo AL
- Subjects
- United States, Humans, National Institute of Allergy and Infectious Diseases (U.S.), Biomarkers, Radiometry, Radioactive Hazard Release, Radiation Injuries prevention & control
- Abstract
Established in 2004, the Radiation and Nuclear Countermeasures Program (RNCP), within the National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health has the central mission to advance medical countermeasure mitigators/therapeutics, and biomarkers and technologies to assess, triage, and inform medical management of patients experiencing acute radiation syndrome and/or the delayed effects of acute radiation exposure. The RNCP biodosimetry mission space encompasses: (1) basic research to elucidate novel approaches for rapid and accurate assessment of radiation exposure, (2) studies to support advanced development for US Food and Drug Administration (FDA) clearance of promising triage or treatment devices/approaches, (3) characterization of biomarkers and/or assays to determine degree of tissue or organ dose that can predict outcome of radiation injuries (i.e., organ failure, morbidity, and/or mortality), and (4) outreach efforts to facilitate interactions with researchers developing cutting edge biodosimetry approaches. Thus far, no biodosimetry device has been FDA cleared for use during a radiological/nuclear incident. At NIAID, advancement of radiation biomarkers and biodosimetry approaches is facilitated by a variety of funding mechanisms (grants, contracts, cooperative and interagency agreements, and Small Business Innovation Research awards), with the objective of advancing devices and assays toward clearance, as outlined in the FDA's Radiation Biodosimetry Medical Countermeasure Devices Guidance. The ultimate goal of the RNCP biodosimetry program is to develop and establish accurate and reliable biodosimetry tools that will improve radiation preparedness and ultimately save lives during a radiological or nuclear incident., (© 2023 The Author(s). Published by S. Karger AG, Basel.)
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.