Network


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

Hotspot


Dive into the research topics where Margaret M. Sedensky is active.

Publication


Featured researches published by Margaret M. Sedensky.


Science | 2013

mTOR Inhibition Alleviates Mitochondrial Disease in a Mouse Model of Leigh Syndrome

Simon C. Johnson; Melana E. Yanos; Ernst Bernhard Kayser; Albert Quintana; Maya Sangesland; Anthony Castanza; Jessica Hui; Valerie Z. Wall; Arni Gagnidze; Kelly Oh; Brian M. Wasko; Fresnida J. Ramos; Richard D. Palmiter; Peter S. Rabinovitch; Philip G. Morgan; Margaret M. Sedensky; Matt Kaeberlein

More from mTOR Leigh syndrome is a rare, untreatable, inherited neurodegenerative disease in children that is caused by functional disruption of mitochondria, the cells energy-producing organelles. Johnson et al. (p. 1524, published online 14 November; see Perspective by Vafai and Mootha) show that rapamycin, a drug used clinically as an immunosuppressant and for treatment of certain cancers, delayed the onset and progression of neurological symptoms in a mouse model of Leigh syndrome and significantly extended survival of the animals. Rapamycin inhibits the so-called “mTOR” signaling pathway, which is currently under intense study because it plays a contributory role in many common diseases. A drug in clinical use for other disorders delays progression of an untreatable mitochondrial disease in knockout mice. [Also see Perspective by Vafai and Mootha] Mitochondrial dysfunction contributes to numerous health problems, including neurological and muscular degeneration, cardiomyopathies, cancer, diabetes, and pathologies of aging. Severe mitochondrial defects can result in childhood disorders such as Leigh syndrome, for which there are no effective therapies. We found that rapamycin, a specific inhibitor of the mechanistic target of rapamycin (mTOR) signaling pathway, robustly enhances survival and attenuates disease progression in a mouse model of Leigh syndrome. Administration of rapamycin to these mice, which are deficient in the mitochondrial respiratory chain subunit Ndufs4 [NADH dehydrogenase (ubiquinone) Fe-S protein 4], delays onset of neurological symptoms, reduces neuroinflammation, and prevents brain lesions. Although the precise mechanism of rescue remains to be determined, rapamycin induces a metabolic shift toward amino acid catabolism and away from glycolysis, alleviating the buildup of glycolytic intermediates. This therapeutic strategy may prove relevant for a broad range of mitochondrial diseases.


PLOS ONE | 2012

Altered anesthetic sensitivity of mice lacking Ndufs4, a subunit of mitochondrial complex I.

Albert Quintana; Philip G. Morgan; Shane E. Kruse; Richard D. Palmiter; Margaret M. Sedensky

Anesthetics are in routine use, yet the mechanisms underlying their function are incompletely understood. Studies in vitro demonstrate that both GABAA and NMDA receptors are modulated by anesthetics, but whole animal models have not supported the role of these receptors as sole effectors of general anesthesia. Findings in C. elegans and in children reveal that defects in mitochondrial complex I can cause hypersensitivity to volatile anesthetics. Here, we tested a knockout (KO) mouse with reduced complex I function due to inactivation of the Ndufs4 gene, which encodes one of the subunits of complex I. We tested these KO mice with two volatile and two non-volatile anesthetics. KO and wild-type (WT) mice were anesthetized with isoflurane, halothane, propofol or ketamine at post-natal (PN) days 23 to 27, and tested for loss of response to tail clamp (isoflurane and halothane) or loss of righting reflex (propofol and ketamine). KO mice were 2.5 - to 3-fold more sensitive to isoflurane and halothane than WT mice. KO mice were 2-fold more sensitive to propofol but resistant to ketamine. These changes in anesthetic sensitivity are the largest recorded in a mammal.


Journal of Cerebral Blood Flow and Metabolism | 2014

Propofol Compared with Isoflurane Inhibits Mitochondrial Metabolism in Immature Swine Cerebral Cortex

Masaki Kajimoto; Douglas B. Atkinson; Dolena R. Ledee; Ernst Bernhard Kayser; Phil G. Morgan; Margaret M. Sedensky; Nancy G. Isern; Christine Des Rosiers; Michael A. Portman

Anesthetics used in infants and children are implicated in the development of neurocognitive disorders. Although propofol induces neuroapoptosis in developing brain, the underlying mechanisms require elucidation and may have an energetic basis. We studied substrate utilization in immature swine anesthetized with either propofol or isoflurane for 4 hours. Piglets were infused with 13-Carbon-labeled glucose and leucine in the common carotid artery to assess citric acid cycle (CAC) metabolism in the parietal cortex. The anesthetics produced similar systemic hemodynamics and cerebral oxygen saturation by near-infrared spectroscopy. Compared with isoflurane, propofol depleted ATP and glycogen stores. Propofol decreased pools of the CAC intermediates, citrate, and α-ketoglutarate, while markedly increasing succinate along with decreasing mitochondrial complex II activity. Propofol also inhibited acetyl-CoA entry into the CAC through pyruvate dehydrogenase, while promoting glycolytic flux with marked lactate accumulation. Although oxygen supply appeared similar between the anesthetic groups, propofol yielded a metabolic phenotype that resembled a hypoxic state. Propofol impairs substrate flux through the CAC in the immature cerebral cortex. These impairments occurred without systemic metabolic perturbations that typically accompany propofol infusion syndrome. These metabolic abnormalities may have a role in the neurotoxity observed with propofol in the vulnerable immature brain.


Disease Models & Mechanisms | 2014

A Drosophila model of mitochondrial disease caused by a complex I mutation that uncouples proton pumping from electron transfer

Jonathon L. Burman; Leslie S. Itsara; Ernst Bernhard Kayser; Wichit Suthammarak; Adrienne M. Wang; Matt Kaeberlein; Margaret M. Sedensky; Philip G. Morgan; Leo J. Pallanck

Mutations affecting mitochondrial complex I, a multi-subunit assembly that couples electron transfer to proton pumping, are the most frequent cause of heritable mitochondrial diseases. However, the mechanisms by which complex I dysfunction results in disease remain unclear. Here, we describe a Drosophila model of complex I deficiency caused by a homoplasmic mutation in the mitochondrial-DNA-encoded NADH dehydrogenase subunit 2 (ND2) gene. We show that ND2 mutants exhibit phenotypes that resemble symptoms of mitochondrial disease, including shortened lifespan, progressive neurodegeneration, diminished neural mitochondrial membrane potential and lower levels of neural ATP. Our biochemical studies of ND2 mutants reveal that complex I is unable to efficiently couple electron transfer to proton pumping. Thus, our study provides evidence that the ND2 subunit participates directly in the proton pumping mechanism of complex I. Together, our findings support the model that diminished respiratory chain activity, and consequent energy deficiency, are responsible for the pathogenesis of complex-I-associated neurodegeneration.


Current Biology | 2016

Glutamatergic Neurotransmission Links Sensitivity to Volatile Anesthetics with Mitochondrial Function

Pavel Zimin; Christian B. Woods; Albert Quintana; Jan-Marino Ramirez; Philip G. Morgan; Margaret M. Sedensky

An enigma of modern medicine has persisted for over 150 years. The mechanisms by which volatile anesthetics (VAs) produce their effects (loss of consciousness, analgesia, amnesia, and immobility) remain an unsolved mystery. Many attractive putative molecular targets have failed to produce a significant effect when genetically tested in whole-animal models [1-3]. However, mitochondrial defects increase VA sensitivity in diverse organisms from nematodes to humans [4-6]. Ndufs4 knockout (KO) mice lack a subunit of mitochondrial complex I and are strikingly hypersensitive to VAs yet resistant to the intravenous anesthetic ketamine [7]. The change in VA sensitivity is the largest reported for a mammal. Limiting NDUFS4 loss to a subset of glutamatergic neurons recapitulates the VA hypersensitivity of Ndufs4(KO) mice, while loss in GABAergic or cholinergic neurons does not. Baseline electrophysiologic function of CA1 pyramidal neurons does not differ between Ndufs4(KO) and control mice. Isoflurane concentrations that anesthetize only Ndufs4(KO) mice (0.6%) decreased the frequency of spontaneous excitatory postsynaptic currents (sEPSCs) only in Ndufs4(KO) CA1 neurons, while concentrations effective in control mice (1.2%) decreased sEPSC frequencies in both control and Ndufs4(KO) CA1 pyramidal cells. Spontaneous inhibitory postsynaptic currents (sIPSCs) were not differentially affected between genotypes. The effects of isoflurane were similar on evoked field excitatory postsynaptic potentials (fEPSPs) and paired pulse facilitation (PPF) in KO and control hippocampal slices. We propose that CA1 presynaptic excitatory neurotransmission is hypersensitive to isoflurane in Ndufs4(KO) mice due to the inhibition of pre-existing reduced complex I function, reaching a critical reduction that can no longer meet metabolic demands.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Tether mutations that restore function and suppress pleiotropic phenotypes of the C. elegans isp-1(qm150) Rieske iron–sulfur protein

Gholamali Jafari; Brian M. Wasko; Ashley Tonge; Nathan Schurman; Cindy Dong; Zhongyu Li; Rebecca Peters; Ernst Bernhard Kayser; Jason N. Pitt; Phil G. Morgan; Margaret M. Sedensky; Antony R. Crofts; Matt Kaeberlein

Significance Mitochondrial function is critical for health and longevity. Mutation of the highly conserved Rieske iron–sulfur subunit (ISP-1) of complex III in the respiratory chain results in pleiotropic phenotypes in Caenorhabditis elegans, including delayed development and increased lifespan. We identified intragenic mutations within a conserved 6-aa tether region of ISP-1. These suppressors are capable of suppressing all of the phenotypes associated with the isp-1(qm150) mutation. We further demonstrated that this mutation/suppressor relationship is conserved in the Rieske iron–sulfur protein (Rip1) of yeast complex III. These findings provide insights into conserved features of the structure and function of this protein, and allow us to propose a unique “spring-loaded” mechanism to account for these effects, supported by empirical physicochemical data. Mitochondria play an important role in numerous diseases as well as normative aging. Severe reduction in mitochondrial function contributes to childhood disorders such as Leigh Syndrome, whereas mild disruption can extend the lifespan of model organisms. The Caenorhabditis elegans isp-1 gene encodes the Rieske iron–sulfur protein subunit of cytochrome c oxidoreductase (complex III of the electron transport chain). The partial loss of function allele, isp-1(qm150), leads to several pleiotropic phenotypes. To better understand the molecular mechanisms of ISP-1 function, we sought to identify genetic suppressors of the delayed development of isp-1(qm150) animals. Here we report a series of intragenic suppressors, all located within a highly conserved six amino acid tether region of ISP-1. These intragenic mutations suppress all of the evaluated isp-1(qm150) phenotypes, including developmental rate, pharyngeal pumping rate, brood size, body movement, activation of the mitochondrial unfolded protein response reporter, CO2 production, mitochondrial oxidative phosphorylation, and lifespan extension. Furthermore, analogous mutations show a similar effect when engineered into the budding yeast Rieske iron–sulfur protein Rip1, revealing remarkable conservation of the structure–function relationship of these residues across highly divergent species. The focus on a single subunit as causal both in generation and in suppression of diverse pleiotropic phenotypes points to a common underlying molecular mechanism, for which we propose a “spring-loaded” model. These observations provide insights into how gating and control processes influence the function of ISP-1 in mediating pleiotropic phenotypes including developmental rate, movement, sensitivity to stress, and longevity.


Genetics | 2017

Cell Biology of the Mitochondrion.

Alexander M. van der Bliek; Margaret M. Sedensky; Phil G. Morgan

Mitochondria are best known for harboring pathways involved in ATP synthesis through the tricarboxylic acid cycle and oxidative phosphorylation. Major advances in understanding these roles were made with Caenorhabditis elegans mutants affecting key components of the metabolic pathways. These mutants have not only helped elucidate some of the intricacies of metabolism pathways, but they have also served as jumping off points for pharmacology, toxicology, and aging studies. The field of mitochondria research has also undergone a renaissance, with the increased appreciation of the role of mitochondria in cell processes other than energy production. Here, we focus on discoveries that were made using C. elegans, with a few excursions into areas that were studied more thoroughly in other organisms, like mitochondrial protein import in yeast. Advances in mitochondrial biogenesis and membrane dynamics were made through the discoveries of novel functions in mitochondrial fission and fusion proteins. Some of these functions were only apparent through the use of diverse model systems, such as C. elegans. Studies of stress responses, exemplified by mitophagy and the mitochondrial unfolded protein response, have also benefitted greatly from the use of model organisms. Recent developments include the discoveries in C. elegans of cell autonomous and nonautonomous pathways controlling the mitochondrial unfolded protein response, as well as mechanisms for degradation of paternal mitochondria after fertilization. The evolutionary conservation of many, if not all, of these pathways ensures that results obtained with C. elegans are equally applicable to studies of human mitochondria in health and disease.


Neurotoxicology and Teratology | 2017

The genetics of isoflurane-induced developmental neurotoxicity.

Hyo-Seok Na; Nicole L Brockway; Katherine Ruth Gentry; Elyce Opheim; Margaret M. Sedensky; Philip G. Morgan

INTRODUCTION Neurotoxicity induced by early developmental exposure to volatile anesthetics is a characteristic of organisms across a wide range of species, extending from the nematode C. elegans to mammals. Prevention of anesthetic-induced neurotoxicity (AIN) will rely upon an understanding of its underlying mechanisms. However, no forward genetic screens have been undertaken to identify the critical pathways affected in AIN. By characterizing such pathways, we may identify mechanisms to eliminate isoflurane induced AIN in mammals. METHODS Chemotaxis in adult C. elegans after larval exposure to isoflurane was used to measure AIN. We initially compared changes in chemotaxis indices between classical mutants known to affect nervous system development adding mutants in response to data. Activation of specific genes was visualized using fluorescent markers. Animals were then treated with rapamycin or preconditioned with isoflurane to test effects on AIN. RESULTS Forty-four mutations, as well as pharmacologic manipulations, identified two pathways, highly conserved from invertebrates to humans, that regulate AIN in C. elegans. Activation of one stress-protective pathway (DAF-2 dependent) eliminates AIN, while activation of a second stress-responsive pathway (endoplasmic reticulum (ER) associated stress) causes AIN. Pharmacologic inhibition of the mechanistic Target of Rapamycin (mTOR) blocks ER-stress and AIN. Preconditioning with isoflurane prior to larval exposure also inhibited AIN. DISCUSSION Our data are best explained by a model in which isoflurane acutely inhibits mitochondrial function causing activation of responses that ultimately lead to ER-stress. The neurotoxic effect of isoflurane can be completely prevented by manipulations at multiple points in the pathways that control this response. Endogenous signaling pathways can be recruited to protect organisms from the neurotoxic effects of isoflurane.


Anesthesiology | 2013

The Power of Unbiased Genetic Screens to Discover Novel Anesthetic Targets

Phil G. Morgan; Margaret M. Sedensky

To the Editor: We are writing in response to Dr. Forman’s1 editorial “The Expanding Genetic Toolkit for Exploring Mechanisms of General Anesthesia” in the April issue of ANESTHESIOLOGY. Dr. Forman covers many excellent points about the use of genetics in understanding anesthetic mechanisms. However, we think that he has overlooked, and perhaps unintentionally discounted, the key ability of an unbiased forward genetic screen to study anesthetic action. A forward screen generates mutations randomly and then looks for those mutations that affect a particular trait. Its unique beauty or power is that it can discover novel mechanisms that would not be found if one presupposed to know an anesthetic target. Forward genetic screens have identified plausible possible targets of volatile anesthetics. They have included leak channels,2 neurotransmitter release machinery,3 and mitochondria.4 All three possibilities have been occurs frequently after the acute phase of trauma-induced coagulopathy.14,15 Dr. Flores suggests the use of multilevel continuous intercostal nerve block catheter in a patient with flail chest. Although the risk of epidural hematoma may be lower with intercostal nerve blocks compared with epidural analgesia, other risks such as pneumothorax/hematothorax and inadequate efficacy may limit its use under the condition described in our case scenario. We need to develop specific outcome-oriented clinical pathways in critical care medicine that do not exclusively take into account the data taken from elective surgical procedures in the operating room. In patients with flail chest presenting with traditional contraindications for neuraxial analgesia, careful risk–benefit analysis may indicate that epidural analgesia improves important outcome measures. We believe that thromboelastography or thromboelastometry and aggregometry (if available) are helpful instruments for decision support in such a case scenario.


Archive | 2018

Using Caenorhabditis elegans to Study Neurotoxicity

Margaret M. Sedensky; Philip G. Morgan

Abstract Studies of neurotoxicity from environmental exposures are difficult to carry out in mammals, with limited genetic tools to sort out underlying mechanisms. However the simple nematode Caenorhabditis elegans has proven a valuable tool to study neurotoxicity. It is an economical and powerful model organism for which the entire genome has been sequenced; the majority of human genes have well-documented orthologs in the worm. Due to decades of genetic studies, classical mutations or interference RNA can be used to interrogate the function of virtually any gene within days. With its short generation time, optical transparency, and array of molecular tools, C. elegans has been widely exploited to study the effects of neurotoxins. In particular the study of manganese, selenium, arsenic, acrylamide, mercury, nicotine, and volatile anesthetics have been explored in this invertebrate. We summarize the recent work exploiting C. elegans to understand neurotoxic effects of these agents.

Collaboration


Dive into the Margaret M. Sedensky's collaboration.

Top Co-Authors

Avatar

Philip G. Morgan

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Phil G. Morgan

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Ernst Bernhard Kayser

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pavel Zimin

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar

Brian M. Wasko

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Ernst-Bernhard Kayser

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sihoun Hahn

Seattle Children's Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge