Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Circ Res ; 132(11): e171-e187, 2023 05 26.
Article En | MEDLINE | ID: mdl-37057625

BACKGROUND: Cardiac contractile function requires high energy from mitochondria, and Ca2+ from the sarcoplasmic reticulum (SR). Via local Ca2+ transfer at close mitochondria-SR contacts, cardiac excitation feedforward regulates mitochondrial ATP production to match surges in demand (excitation-bioenergetics coupling). However, pathological stresses may cause mitochondrial Ca2+ overload, excessive reactive oxygen species production and permeability transition, risking homeostatic collapse and myocyte loss. Excitation-bioenergetics coupling involves mitochondria-SR tethers but the role of tethering in cardiac physiology/pathology is debated. Endogenous tether proteins are multifunctional; therefore, nonselective targets to scrutinize interorganelle linkage. Here, we assessed the physiological/pathological relevance of selective chronic enhancement of cardiac mitochondria-SR tethering. METHODS: We introduced to mice a cardiac muscle-specific engineered tether (linker) transgene with a fluorescent protein core and deployed 2D/3D electron microscopy, biochemical approaches, fluorescence imaging, in vivo and ex vivo cardiac performance monitoring and stress challenges to characterize the linker phenotype. RESULTS: Expressed in the mature cardiomyocytes, the linker expanded and tightened individual mitochondria-junctional SR contacts; but also evoked a marked remodeling with large dense mitochondrial clusters that excluded dyads. Yet, excitation-bioenergetics coupling remained well-preserved, likely due to more longitudinal mitochondria-dyad contacts and nanotunnelling between mitochondria exposed to junctional SR and those sealed away from junctional SR. Remarkably, the linker decreased female vulnerability to acute massive ß-adrenergic stress. It also reduced myocyte death and mitochondrial calcium-overload-associated myocardial impairment in ex vivo ischemia/reperfusion injury. CONCLUSIONS: We propose that mitochondria-SR/endoplasmic reticulum contacts operate at a structural optimum. Although acute changes in tethering may cause dysfunction, upon chronic enhancement of contacts from early life, adaptive remodeling of the organelles shifts the system to a new, stable structural optimum. This remodeling balances the individually enhanced mitochondrion-junctional SR crosstalk and excitation-bioenergetics coupling, by increasing the connected mitochondrial pool and, presumably, Ca2+/reactive oxygen species capacity, which then improves the resilience to stresses associated with dysregulated hyperactive Ca2+ signaling.


Calcium Signaling , Sarcoplasmic Reticulum , Female , Mice , Animals , Sarcoplasmic Reticulum/metabolism , Reactive Oxygen Species/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Mitochondria, Heart/metabolism , Calcium/metabolism
2.
FASEB J ; 35(12): e21999, 2021 12.
Article En | MEDLINE | ID: mdl-34748223

The Creb-Regulated Transcriptional Coactivator (Crtc) family of transcriptional coregulators drive Creb1-mediated transcription effects on metabolism in many tissues, but the in vivo effects of Crtc2/Creb1 transcription on skeletal muscle metabolism are not known. Skeletal muscle-specific overexpression of Crtc2 (Crtc2 mice) induced greater mitochondrial activity, metabolic flux capacity for both carbohydrates and fats, improved glucose tolerance and insulin sensitivity, and increased oxidative capacity, supported by upregulation of key metabolic genes. Crtc2 overexpression led to greater weight loss during alternate day fasting (ADF), selective loss of fat rather than lean mass, maintenance of higher energy expenditure during the fast and reduced binge-eating during the feeding period. ADF downregulated most of the mitochondrial electron transport genes, and other regulators of mitochondrial function, that were substantially reversed by Crtc2-driven transcription. Glucocorticoids acted with AMPK to drive atrophy and mitophagy, which was reversed by Crtc2/Creb1 signaling. Crtc2/Creb1-mediated signaling coordinates metabolic adaptations in skeletal muscle that explain how Crtc2/Creb1 regulates metabolism and weight loss.


Cyclic AMP Response Element-Binding Protein/physiology , Energy Metabolism , Fasting , Insulin Resistance , Muscle, Skeletal/physiology , Transcription Factors/physiology , Weight Loss/physiology , Animals , Male , Mice , Mice, Transgenic
3.
J Gen Physiol ; 152(11)2020 11 02.
Article En | MEDLINE | ID: mdl-33005936
4.
Cell Death Dis ; 11(8): 661, 2020 08 19.
Article En | MEDLINE | ID: mdl-32814770

The mitochondrial permeability transition pore (mPTP) plays a critical role in the pathogenesis of cardiovascular diseases, including ischemia/reperfusion injury. Although the pore structure is still unresolved, the mechanism through which cyclophilin D (CypD) regulates mPTP opening is the subject of intensive studies. While post-translational modifications of CypD have been shown to modulate pore opening, specific phosphorylation sites of CypD have not yet been identified. We hypothesized here that phosphorylation of CypD on a serine residue controls mPTP opening and subsequent cell death at reperfusion. We combined in silico analysis with in vitro and genetic manipulations to determine potential CypD phosphorylation sites and their effect on mitochondrial function and cell death. Importantly, we developed an in vivo intramyocardial adenoviral strategy to assess the effect of the CypD phosphorylation event on infarct size. Our results show that although CypD can potentially be phosphorylated at multiple serine residues, only the phosphorylation status at S191 directly impacts the ability of CypD to regulate the mPTP. Protein-protein interaction strategies showed that the interaction between CypD and oligomycin sensitivity-conferring protein (OSCP) was reduced by 45% in the phosphoresistant S191A mutant, whereas it was increased by 48% in the phosphomimetic S191E mutant cells. As a result, the phosphoresistant CypD S191A mutant was protected against 18 h starvation whereas cell death was significantly increased in phosphomimetic S191E group, associated with mitochondrial respiration alteration and ROS production. As in vivo proof of concept, in S191A phosphoresistant rescued CypD-KO mice developed significantly smaller infarct as compared to WT whereas infarct size was drastically increased in S191E phosphomimetic rescued mice. We conclude that CypD phosphorylation at S191 residue leads to its binding to OSCP and thus sensitizes mPTP opening for the subsequent cell death.


Mitochondrial Permeability Transition Pore/metabolism , Peptidyl-Prolyl Isomerase F/genetics , Peptidyl-Prolyl Isomerase F/metabolism , Animals , Cell Death , Ischemia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Phosphorylation , Reperfusion , Reperfusion Injury/metabolism , Serine , Signal Transduction/drug effects
5.
J Am Med Inform Assoc ; 27(9): 1456-1461, 2020 07 01.
Article En | MEDLINE | ID: mdl-32618999

The COVID-19 pandemic has led to the rapid expansion of telehealth services as healthcare organizations aim to mitigate community transmission while providing safe patient care. As technology adoption rapidly increases, operational telehealth teams must maintain awareness of critical information, such as patient volumes and wait times, patient and provider experience, and telehealth platform performance. Using a model of situation awareness as a conceptual foundation and a user-centered design approach we describe our process for rapidly developing and disseminating dashboard visualizations to support telehealth operations. We used a 5-step process to gain domain knowledge, identify user needs, identify data sources, design and develop visualizations, and iteratively refine these visualizations. Through this process we identified 3 distinct stakeholder groups and designed and developed visualization dashboards to meet their needs. Feedback from users demonstrated the dashboard's support situation awareness and informed important operational decisions. Lessons learned are shared to provide other organizations with insights from our process.


Coronavirus Infections , Data Display , Data Visualization , Pandemics , Pneumonia, Viral , Telemedicine , Betacoronavirus , COVID-19 , Humans , Mid-Atlantic Region , Multi-Institutional Systems , Organizational Case Studies , SARS-CoV-2 , User-Computer Interface
6.
J Biol Chem ; 294(28): 10807-10818, 2019 07 12.
Article En | MEDLINE | ID: mdl-31097542

The mitochondrial matrix ATPase associated with diverse cellular activities (m-AAA) protease spastic paraplegia 7 (SPG7) has been recently implicated as either a negative or positive regulatory component of the mitochondrial permeability transition pore (mPTP) by two research groups. To address this controversy, we investigated possible mechanisms that explain the discrepancies between these two studies. We found that loss of the SPG7 gene increased resistance to Ca2+-induced mPTP opening. However, this occurs independently of cyclophilin D (cyclosporine A insensitive) rather it is through decreased mitochondrial Ca2+ concentrations and subsequent adaptations mediated by impaired formation of functional mitochondrial Ca2+ uniporter complexes. We found that SPG7 directs the m-AAA complex to favor association with the mitochondrial Ca2+ uniporter (MCU) and MCU processing regulates higher order MCU-complex formation. The results suggest that SPG7 does not constitute a core component of the mPTP but can modulate mPTP through regulation of the basal mitochondrial Ca2+ concentration.


ATPases Associated with Diverse Cellular Activities/metabolism , Calcium Channels/metabolism , Metalloendopeptidases/metabolism , ATPases Associated with Diverse Cellular Activities/physiology , Calcium/metabolism , Cell Membrane Permeability/physiology , HEK293 Cells , Humans , Metalloendopeptidases/physiology , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/physiology , Mitochondrial Membranes/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Transmembrane Permeability-Driven Necrosis/physiology , Paraplegia/metabolism , Proton-Translocating ATPases/metabolism , Spastic Paraplegia, Hereditary/metabolism
7.
Clin Cancer Res ; 23(17): 5238-5245, 2017 Sep 01.
Article En | MEDLINE | ID: mdl-28536307

Purpose: The association of tumor gene expression profiles with progression-free survival (PFS) outcomes in patients with BRAFV600-mutated melanoma treated with vemurafenib or cobimetinib combined with vemurafenib was evaluated.Experimental Design: Gene expression of archival tumor samples from patients in four trials (BRIM-2, BRIM-3, BRIM-7, and coBRIM) was evaluated. Genes significantly associated with PFS (P < 0.05) were identified by univariate Cox proportional hazards modeling, then subjected to unsupervised hierarchical clustering, principal component analysis, and recursive partitioning to develop optimized gene signatures.Results: Forty-six genes were identified as significantly associated with PFS in both BRIM-2 (n = 63) and the vemurafenib arm of BRIM-3 (n = 160). Two distinct signatures were identified: cell cycle and immune. Among vemurafenib-treated patients, the cell-cycle signature was associated with shortened PFS compared with the immune signature in the BRIM-2/BRIM-3 training set [hazard ratio (HR) 1.8; 95% confidence interval (CI), 1.3-2.6, P = 0.0001] and in the coBRIM validation set (n = 101; HR, 1.6; 95% CI, 1.0-2.5; P = 0.08). The adverse impact of the cell-cycle signature on PFS was not observed in patients treated with cobimetinib combined with vemurafenib (n = 99; HR, 1.1; 95% CI, 0.7-1.8; P = 0.66).Conclusions: In vemurafenib-treated patients, the cell-cycle gene signature was associated with shorter PFS. However, in cobimetinib combined with vemurafenib-treated patients, both cell cycle and immune signature subgroups had comparable PFS. Cobimetinib combined with vemurafenib may abrogate the adverse impact of the cell-cycle signature. Clin Cancer Res; 23(17); 5238-45. ©2017 AACR.


Azetidines/administration & dosage , Indoles/administration & dosage , Melanoma/drug therapy , Piperidines/administration & dosage , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Azetidines/adverse effects , Disease-Free Survival , Drug Resistance, Neoplasm/genetics , Female , Humans , Indoles/adverse effects , Male , Melanoma/genetics , Melanoma/pathology , Middle Aged , Mutation , Piperidines/adverse effects , Proportional Hazards Models , Sulfonamides/adverse effects , Treatment Outcome , Vemurafenib
8.
Handb Exp Pharmacol ; 240: 129-156, 2017.
Article En | MEDLINE | ID: mdl-28194521

Mitochondrial Ca2+ uptake is crucial for an array of cellular functions while an imbalance can elicit cell death. In this chapter, we briefly reviewed the various modes of mitochondrial Ca2+ uptake and our current understanding of mitochondrial Ca2+ homeostasis in regards to cell physiology and pathophysiology. Further, this chapter focuses on the molecular identities, intracellular regulators as well as the pharmacology of mitochondrial Ca2+ uniporter complex.


Calcium Channels/physiology , Animals , Calcium/metabolism , Calcium Channels/chemistry , Calcium Channels/drug effects , Energy Metabolism , Homeostasis , Humans , Mitochondria/metabolism
9.
J Bioenerg Biomembr ; 49(1): 27-47, 2017 Feb.
Article En | MEDLINE | ID: mdl-27497945

The mitochondrial permeability transition pore was originally described in the 1970's as a Ca2+ activated pore and has since been attributed to the pathogenesis of many diseases. Here we evaluate how each of the current models of the pore complex fit to what is known about how Ca2+ regulates the pore, and any insight that provides into the molecular identity of the pore complex. We also discuss the central role of Ca2+ in modulating the pore's open probability by directly regulating processes, such as ATP/ADP balance through the tricarboxylic acid cycle, electron transport chain, and mitochondrial membrane potential. We review how Ca2+ influences second messengers such as reactive oxygen/nitrogen species production and polyphosphate formation. We discuss the evidence for how Ca2+ regulates post-translational modification of cyclophilin D including phosphorylation by glycogen synthase kinase 3 beta, deacetylation by sirtuins, and oxidation/ nitrosylation of key residues. Lastly we introduce a novel view into how Ca2+ activated proteolysis through calpains in the mitochondria may be a driver of sustained pore opening during pathologies such as ischemia reperfusion injury.


Calcium/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Animals , Peptidyl-Prolyl Isomerase F , Cyclophilins/metabolism , Humans , Membrane Potential, Mitochondrial , Mitochondria, Heart , Mitochondrial Permeability Transition Pore , Proteolysis , Reactive Oxygen Species/metabolism , Reperfusion Injury
10.
Biochem Biophys Res Commun ; 465(3): 464-70, 2015 Sep 25.
Article En | MEDLINE | ID: mdl-26277396

Protein kinase C (PKC) plays key roles in the regulation of signal transduction and cellular function in various cell types. At least ten PKC isoforms have been identified and intracellular localization and trafficking of these individual isoforms are important for regulation of enzyme activity and substrate specificity. PKC can be activated downstream of Gq-protein coupled receptor (GqPCR) signaling and translocate to various cellular compartments including plasma membrane (PM). Recent reports suggested that different types of GqPCRs would activate different PKC isoforms (classic, novel and atypical PKCs) with different trafficking patterns. However, the knowledge of isoform-specific activation of PKC by each GqPCR is limited. α1-Adrenoceptor (α1-AR) is one of the GqPCRs highly expressed in the cardiovascular system. In this study, we examined the isoform-specific dynamic translocation of PKC in living HEK293T cells by α1-AR stimulation (α1-ARS). Rat PKCα, ßI, ßII, δ, ε and ζ fused with GFP at C-term were co-transfected with human α1A-AR into HEK293T cells. The isoform-specific dynamic translocation of PKC in living HEK293T cells by α1-ARS using phenylephrine was measured by confocal microscopy. Before stimulation, GFP-PKCs were localized at cytosolic region. α1-ARS strongly and rapidly translocated a classical PKC (cPKC), PKCα, (<30 s) to PM, with PKCα returning diffusively into the cytosol within 5 min. α1-ARS rapidly translocated other cPKCs, PKCßI and PKCßII, to the PM (<30 s), with sustained membrane localization. One novel PKC (nPKC), PKCε, but not another nPKC, PKCδ, was translocated by α1-AR stimulation to the PM (<30 s) and its membrane localization was also sustained. Finally, α1-AR stimulation did not cause a diacylglycerol-insensitive atypical PKC, PKCζ translocation. Our data suggest that PKCα, ß and ε activation may underlie physiological and pathophysiological responses of α1-AR signaling for the phosphorylation of membrane-associated substrates including ion-channel and transporter proteins in the cardiovascular system.


Cell Membrane/metabolism , Protein Kinase C/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction/physiology , HEK293 Cells , Humans , Protein Isoforms/metabolism , Protein Transport/physiology
11.
Antioxid Redox Signal ; 21(6): 863-79, 2014 Aug 20.
Article En | MEDLINE | ID: mdl-24800979

AIMS: Mitochondrial Ca2+ homeostasis is crucial for balancing cell survival and death. The recent discovery of the molecular identity of the mitochondrial Ca2+ uniporter pore (MCU) opens new possibilities for applying genetic approaches to study mitochondrial Ca2+ regulation in various cell types, including cardiac myocytes. Basal tyrosine phosphorylation of MCU was reported from mass spectroscopy of human and mouse tissues, but the signaling pathways that regulate mitochondrial Ca2+ entry through posttranslational modifications of MCU are completely unknown. Therefore, we investigated α1-adrenergic-mediated signal transduction of MCU posttranslational modification and function in cardiac cells. RESULTS: α1-adrenoceptor (α1-AR) signaling translocated activated proline-rich tyrosine kinase 2 (Pyk2) from the cytosol to mitochondrial matrix and accelerates mitochondrial Ca2+ uptake via Pyk2-dependent MCU phosphorylation and tetrametric MCU channel pore formation. Moreover, we found that α1-AR stimulation increases reactive oxygen species production at mitochondria, mitochondrial permeability transition pore activity, and initiates apoptotic signaling via Pyk2-dependent MCU activation and mitochondrial Ca2+ overload. INNOVATION: Our data indicate that inhibition of α1-AR-Pyk2-MCU signaling represents a potential novel therapeutic target to limit or prevent mitochondrial Ca2+ overload, oxidative stress, mitochondrial injury, and myocardial death during pathophysiological conditions, where chronic adrenergic stimulation is present. CONCLUSION: The α1-AR-Pyk2-dependent tyrosine phosphorylation of the MCU regulates mitochondrial Ca2+ entry and apoptosis in cardiac cells.


Calcium Channels/metabolism , Calcium/metabolism , Focal Adhesion Kinase 2/metabolism , Mitochondria/metabolism , Signal Transduction , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Line , Cytosol/metabolism , Humans , Models, Biological , Myocytes, Cardiac/metabolism , Phosphorylation , Protein Binding , Protein Multimerization , Protein Transport , Rats , Reactive Oxygen Species/metabolism , Receptors, Adrenergic, alpha-1/metabolism
12.
Neurosci Lett ; 575: 7-12, 2014 Jul 11.
Article En | MEDLINE | ID: mdl-24861510

Mitochondrial Ca(2+) controls numerous cell functions, such as energy metabolism, reactive oxygen species generation, spatiotemporal dynamics of Ca(2+) signaling, cell growth and death in various cell types including neurons. Mitochondrial Ca(2+) accumulation is mainly mediated by the mitochondrial Ca(2+) uniporter (MCU), but recent reports also indicate that mitochondrial Ca(2+)-influx mechanisms are regulated not only by MCU, but also by multiple channels/transporters. We previously reported that ryanodine receptor (RyR), which is a one of the main Ca(2+)-release channels at endoplasmic/sarcoplasmic reticulum (SR/ER) in excitable cells, is expressed at the mitochondrial inner membrane (IMM) and serves as a part of the Ca(2+) uptake mechanism in cardiomyocytes. Although RyR is also expressed in neuronal cells and works as a Ca(2+)-release channel at ER, it has not been well investigated whether neuronal mitochondria possess RyR and, if so, whether this mitochondrial RyR has physiological functions in neuronal cells. Here we show that neuronal mitochondria express RyR at IMM and accumulate Ca(2+) through this channel in response to cytosolic Ca(2+) elevation, which is similar to what we observed in another excitable cell-type, cardiomyocytes. In addition, the RyR blockers dantrolene or ryanodine significantly inhibits mitochondrial Ca(2+) uptake in permeabilized striatal neurons. Taken together, we identify RyR as an additional mitochondrial Ca(2+) uptake mechanism in response to the elevation of [Ca(2+)]c in neurons, suggesting that this channel may play a critical role in mitochondrial Ca(2+)-mediated functions such as energy metabolism.


Mitochondria/metabolism , Neurons/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cells, Cultured , Corpus Striatum/cytology , Dantrolene/pharmacology , Mitochondrial Membranes/metabolism , Rats, Sprague-Dawley , Ryanodine/pharmacology
13.
Antioxid Redox Signal ; 21(6): 987-1006, 2014 Aug 20.
Article En | MEDLINE | ID: mdl-24180309

SIGNIFICANCE: Mitochondrial ion channels/transporters and the electron transport chain (ETC) serve as key sensors and regulators for cellular redox signaling, the production of reactive oxygen species (ROS) and nitrogen species (RNS) in mitochondria, and balancing cell survival and death. Although the functional and pharmacological characteristics of mitochondrial ion transport mechanisms have been extensively studied for several decades, the majority of the molecular identities that are responsible for these channels/transporters have remained a mystery until very recently. RECENT ADVANCES: Recent breakthrough studies uncovered the molecular identities of the diverse array of major mitochondrial ion channels/transporters, including the mitochondrial Ca2+ uniporter pore, mitochondrial permeability transition pore, and mitochondrial ATP-sensitive K+ channel. This new information enables us to form detailed molecular and functional characterizations of mitochondrial ion channels/transporters and their roles in mitochondrial redox signaling. CRITICAL ISSUES: Redox-mediated post-translational modifications of mitochondrial ion channels/transporters and ETC serve as key mechanisms for the spatiotemporal control of mitochondrial ROS/RNS generation. FUTURE DIRECTIONS: Identification of detailed molecular mechanisms for redox-mediated regulation of mitochondrial ion channels will enable us to find novel therapeutic targets for many diseases that are associated with cellular redox signaling and mitochondrial ion channels/transporters.


Ion Channels/metabolism , Mitochondria/metabolism , Signal Transduction , Animals , Calcium/metabolism , Electron Transport Chain Complex Proteins/metabolism , Humans , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Permeability Transition Pore , Oxidation-Reduction , Potassium Channels/metabolism , Reactive Oxygen Species/metabolism , Voltage-Dependent Anion Channels/metabolism
14.
Am J Physiol Heart Circ Physiol ; 305(12): H1736-51, 2013 Dec.
Article En | MEDLINE | ID: mdl-24124188

Ca(+) influx to mitochondria is an important trigger for both mitochondrial dynamics and ATP generation in various cell types, including cardiac cells. Mitochondrial Ca(2+) influx is mainly mediated by the mitochondrial Ca(2+) uniporter (MCU). Growing evidence also indicates that mitochondrial Ca(2+) influx mechanisms are regulated not solely by MCU but also by multiple channels/transporters. We have previously reported that skeletal muscle-type ryanodine receptor (RyR) type 1 (RyR1), which expressed at the mitochondrial inner membrane, serves as an additional Ca(2+) uptake pathway in cardiomyocytes. However, it is still unclear which mitochondrial Ca(2+) influx mechanism is the dominant regulator of mitochondrial morphology/dynamics and energetics in cardiomyocytes. To investigate the role of mitochondrial RyR1 in the regulation of mitochondrial morphology/function in cardiac cells, RyR1 was transiently or stably overexpressed in cardiac H9c2 myoblasts. We found that overexpressed RyR1 was partially localized in mitochondria as observed using both immunoblots of mitochondrial fractionation and confocal microscopy, whereas RyR2, the main RyR isoform in the cardiac sarcoplasmic reticulum, did not show any expression at mitochondria. Interestingly, overexpression of RyR1 but not MCU or RyR2 resulted in mitochondrial fragmentation. These fragmented mitochondria showed bigger and sustained mitochondrial Ca(2+) transients compared with basal tubular mitochondria. In addition, RyR1-overexpressing cells had a higher mitochondrial ATP concentration under basal conditions and showed more ATP production in response to cytosolic Ca(2+) elevation compared with nontransfected cells as observed by a matrix-targeted ATP biosensor. These results indicate that RyR1 possesses a mitochondrial targeting/retention signal and modulates mitochondrial morphology and Ca(2+)-induced ATP production in cardiac H9c2 myoblasts.


Adenosine Triphosphate/biosynthesis , Calcium/metabolism , Mitochondria/metabolism , Myoblasts, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium Signaling/physiology , Cell Line , Mitochondria/genetics , Rats , Ryanodine Receptor Calcium Release Channel/genetics
15.
Biochem Biophys Res Commun ; 433(2): 188-93, 2013 Apr 05.
Article En | MEDLINE | ID: mdl-23454381

Adrenoceptor stimulation is a key determinant of cardiac excitation-contraction coupling mainly through the activation of serine/threonine kinases. However, little is known about the role of protein tyrosine kinases (PTKs) activated by adrenergic signaling on cardiac excitation-contraction coupling. A cytoplasmic tyrosine residue in ß1-adrenoceptor is estimated to regulate Gs-protein binding affinity from crystal structure studies, but the signaling pathway leading to the phosphorylation of these residues is unknown. Here we show α1-adrenergic signaling inhibits ß-adrenergically activated Ca(2+) current, Ca(2+) transients and contractile force through phosphorylation of tyrosine residues in ß1-adrenoceptor by PTK. Our results indicate that inhibition of ß-adrenoceptor-mediated Ca(2+) elevation by α1-adrenoceptor-PTK signaling serves as an important regulatory feedback mechanism when the catecholamine level increases to protect cardiomyocytes from cytosolic Ca(2+) overload.


Adrenergic alpha-1 Receptor Agonists/pharmacology , Excitation Contraction Coupling/drug effects , Papillary Muscles/drug effects , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Adrenergic, beta-1/metabolism , Tyrosine/metabolism , Adenylyl Cyclases/metabolism , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Calcium Signaling/drug effects , Cytosol/metabolism , Heart Ventricles/drug effects , Humans , In Vitro Techniques , Isoproterenol/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Papillary Muscles/physiology , Patch-Clamp Techniques , Phenylephrine/pharmacology , Phosphorylation , Propanolamines/pharmacology , Rats
16.
HERD ; 5(3): 46-65, 2012.
Article En | MEDLINE | ID: mdl-23002568

OBJECTIVE: The aim of this pilot study was to test design interventions such as lighting, color, and spatial color patterning on nurses' stress, alertness, and satisfaction, and to provide an example of how clinical simulation centers can be used to conduct research. BACKGROUND: The application of evidence-based design research in healthcare settings requires a transdisciplinary approach. Integrating approaches from multiple fields in real-life settings often proves time consuming and experimentally difficult. However, forums for collaboration such as clinical simulation centers may offer a solution. In these settings, identical operating and patient rooms are used to deliver simulated patient care scenarios using automated mannequins. METHODS: Two identical rooms were modified in the clinical simulation center. Nurses spent 30 minutes in each room performing simulated cardiac resuscitation. Subjective measures of nurses' stress, alertness, and satisfaction were collected and compared between settings and across time using matched-pair t-test analysis. RESULTS: Nurses reported feeling less stressed after exposure to the experimental room than nurses who were exposed to the control room (2.22, p = .03). Scores post-session indicated a significant reduction in stress and an increase in alertness after exposure to the experimental room as compared to the control room, with significance levels below .10. (Change in stress scores: 3.44, p = .069); (change in alertness scores: 3.6, p = .071). CONCLUSION: This study reinforces the use of validated survey tools to measure stress, alertness, and satisfaction. Results support human-centered design approaches by evaluating the effect on nurses in an experimental setting.


Color , Interior Design and Furnishings/methods , Lighting , Nursing Staff, Hospital/psychology , Adult , Behavioral Risk Factor Surveillance System , Evidence-Based Practice , Female , Focus Groups , Humans , Job Satisfaction , Pilot Projects , Stress, Psychological
17.
Hosp Top ; 90(3): 57-64, 2012.
Article En | MEDLINE | ID: mdl-22989223

Modern medical simulation technology (MST) debuted in 1960 with the development of Resusci Annie (Laerdal 2007), which assisted students in the acquisition of proper ventilation and compression techniques used during basic life support. Following a steady stream of subsequent technological advances and innovations, MST manufacturers are now able to offer training aids capable of facilitating innovative learning in such diverse areas as human patient simulators, simulated clinical environments, virtual procedure stations, virtual medical environments, electronic tutors, and performance recording. The authors list a number of the most popular MSTs presently available while citing evaluative efforts undertaken to date regarding the efficacy of MST to the medical profession. They conclude by proposing a variety of simulation innovations of prospective interest to both medical and technology personnel while offering healthcare administrators a series of recommended considerations when planning to integrate MST into existing medical systems.


Education, Medical/methods , Technology , Clinical Competence , Computer-Assisted Instruction , General Surgery/education , Humans , Patient Simulation , Program Evaluation , Technology/economics , User-Computer Interface
19.
J Exp Med ; 204(1): 161-70, 2007 Jan 22.
Article En | MEDLINE | ID: mdl-17200411

Interleukin (IL)-25 is a member of the IL-17 family of cytokines. However, unlike the other members of this family, IL-25 promotes T helper (Th) 2 responses. We now show that IL-25 also regulates the development of autoimmune inflammation mediated by IL-17-producing T cells. We have generated IL-25-deficient (il25-/-) mice and found that they are highly susceptible to experimental autoimmune encephalomyelitis (EAE). The accelerated disease in the il25-/- mice is associated with an increase of IL-23 in the periphery and a subsequent increase in the number of inflammatory IL-17-, IFNgamma-, and TNF-producing T cells that invade the central nervous system. Neutralization of IL-17 but not IFNgamma in il25-/- mice prevented EAE, suggesting that IL-17 is a major disease-promoting factor. IL-25 treatment at several time points during a relapse-remitting model or chronic model of EAE completely suppressed disease. IL-25 treatment induced elevated production of IL-13, which is required for suppression of Th17 responses by direct inhibition of IL-23, IL-1beta, and IL-6 expression in activated dendritic cells. Thus, IL-25 and IL-17, being members of the same cytokine family, play opposing roles in the pathogenesis of organ-specific autoimmunity.


Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/metabolism , Interleukins/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Animals , Autoimmunity , Base Sequence , Central Nervous System/immunology , DNA/genetics , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Inflammation/etiology , Inflammation/immunology , Interferon-gamma/biosynthesis , Interleukins/deficiency , Interleukins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Th2 Cells/immunology
20.
Cell ; 123(5): 931-44, 2005 Dec 02.
Article En | MEDLINE | ID: mdl-16325585

Innate immunity is the first line of defense against infection, protecting the host during the development of adaptive immunity and critically affecting the nature of the adaptive response. We show that, in contrast to tumor necrosis factor alpha (TNF-alpha), the related protein TWEAK attenuates the transition from innate to adaptive mechanisms. TWEAK-/- mice had overabundant natural killer (NK) cells and displayed hypersensitivity to bacterial endotoxin, with their innate immune cells producing excess interferon (IFN)-gamma and interleukin (IL)-12. TWEAK inhibited stimulation of the transcriptional activator STAT-1 and induced p65 nuclear factor (NF)-kappaB association with histone deacetylase 1, repressing cytokine production. TWEAK-/- mice developed oversized spleens with expanded memory and T helper 1 (TH1) subtype cells upon aging and mounted stronger innate and adaptive TH1-based responses against tumor challenge. Thus, TWEAK suppresses production of IFN-gamma and IL-12, curtailing the innate response and its transition to adaptive TH1 immunity.


Immunity, Cellular/physiology , Immunity, Innate/physiology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factors/immunology , Animals , Cells, Cultured , Cytokine TWEAK , Endotoxins/immunology , Histone Deacetylase 1 , Histone Deacetylases/metabolism , Humans , Hypersensitivity/immunology , Interferon-gamma/immunology , Interleukin-12/immunology , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Mice , Mice, Knockout , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , STAT1 Transcription Factor/metabolism , Spleen/anatomy & histology , Spleen/immunology , T-Lymphocyte Subsets/immunology , TWEAK Receptor , Transcription Factor RelA/metabolism , Tumor Necrosis Factors/genetics
...