626 results on '"Rutter, Jared"'
Search Results
252. Bach, C.P.E.: Concertos for harpsichord and strings in A major and in D major
- Author
-
Rutter, Jared
- Subjects
Bach, C.P.E.: Concertos for Harpsichord and Strings in A Major and in D Major (Sound recording) -- Sound recording reviews ,Sound recordings ,Anthropology/archeology/folklore ,History - Published
- 1981
253. La Mantovana
- Author
-
Rutter, Jared
- Subjects
La Mantovana (Sound recording) -- Sound recording reviews ,Sound recordings ,Anthropology/archeology/folklore ,History - Published
- 1981
254. Renaissance brass
- Author
-
Rutter, Jared
- Subjects
Renaissance Brass (Sound recording) -- Sound recording reviews ,Sound recordings ,Anthropology/archeology/folklore ,History - Published
- 1981
255. American brass band journal revisited
- Author
-
Rutter, Jared
- Subjects
American Brass Band Journal Revisited (Sound recording) -- Sound recording reviews ,Sound recordings ,Anthropology/archeology/folklore ,History - Published
- 1981
256. Tudor women
- Author
-
Rutter, Jared
- Subjects
Tudor Women (Book) -- Book reviews ,Books ,Anthropology/archeology/folklore ,History - Published
- 1979
257. Mozart: Rondo in A minor; Rondo in D major; Beethoven: Sonata No. 2 ('Moonlight')
- Author
-
Rutter, Jared
- Subjects
Mozart: Rondo in A Minor; Rondo in D Major; Beethoven: Sonata No. 2 ('Moonlight') (Sound recording) -- Sound recording reviews ,Sound recordings ,Anthropology/archeology/folklore ,History - Published
- 1981
258. A palace for a king
- Author
-
Rutter, Jared
- Subjects
A Palace For A King (Book) -- Book reviews ,Books ,Anthropology/archeology/folklore ,History - Published
- 1981
259. Haydn
- Author
-
Rutter, Jared
- Subjects
Haydn (Book) -- Book reviews ,Books ,Anthropology/archeology/folklore ,History - Published
- 1979
260. The unknown Weill
- Author
-
Rutter, Jared
- Subjects
The Unknown Weill (Sound recording) -- Sound recording reviews ,Sound recordings ,Anthropology/archeology/folklore ,History - Published
- 1982
261. Lieder
- Author
-
Rutter, Jared
- Subjects
Lieder (Sound recording) -- Sound recording reviews ,Sound recordings ,Anthropology/archeology/folklore ,History - Published
- 1982
262. On the cutting edge: perspectives in bioenergetics.
- Author
-
Granath-Panelo, Melia, Krook, Anna, Rutter, Jared, and Kajimura, Shingo
- Subjects
- *
BIOENERGETICS , *ADIPOSE tissues , *DRUG target - Abstract
The field of bioenergetics is rapidly expanding with new discoveries of mechanisms and potential therapeutic targets. The 2023 Keystone symposium on 'Bioenergetics in Health and Disease', which was jointly held with the symposium 'Adipose Tissue: Energizing Good Fat', consisted of a powerhouse line-up of researchers who shared their insights. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
263. Revealing human disease genes through analysis of the yeast mitochondrial proteome.
- Author
-
Huai-Xiang Hao and Rutter, Jared
- Published
- 2009
- Full Text
- View/download PDF
264. Maestro of the SereNADe: SLC25A51 Orchestrates Mitochondrial NAD+.
- Author
-
Ouyang, Yeyun, Bott, Alex J., and Rutter, Jared
- Subjects
- *
MITOCHONDRIA , *NAD (Coenzyme) , *BIOLOGY , *RESPIRATION , *NICOTINAMIDE - Abstract
Recently, three groups, Girardi et al. , Kory et al. , and Luongo et al. , independently identified solute carrier (SLC) 25A51 as the long-sought, major mitochondrial NAD+ transporter in mammalian cells. These studies not only deorphan an uncharacterized transporter of the SLC25A family, but also shed light on other aspects of NAD+ biology. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
265. Transferred mitochondria accumulate reactive oxygen species, promoting proliferation.
- Author
-
Kidwell, Chelsea U., Casalini, Joseph R., Pradeep, Soorya, Scherer, Sandra D., Greiner, Daniel, Bayik, Defne, Watson, Dionysios C., Olson, Gregory S., Lathia, Justin D., Johnson, Jarrod S., Rutter, Jared, Welm, Alana L., Zangle, Thomas A., and Roh-Johnson, Minna
- Subjects
- *
REACTIVE oxygen species , *CELL physiology , *PLANT mitochondria , *MITOCHONDRIA , *CANCER cell proliferation , *BIOENERGETICS - Abstract
Recent studies reveal that lateral mitochondrial transfer, the movement of mitochondria from one cell to another, can affect cellular and tissue homeostasis. Most of what we know about mitochondrial transfer stems from bulk cell studies and have led to the paradigm that functional transferred mitochondria restore bioenergetics and revitalize cellular functions to recipient cells with damaged or non-functional mitochondrial networks. However, we show that mitochondrial transfer also occurs between cells with functioning endogenous mitochondrial networks, but the mechanisms underlying how transferred mitochondria can promote such sustained behavioral reprogramming remain unclear. We report that unexpectedly, transferred macrophage mitochondria are dysfunctional and accumulate reactive oxygen species in recipient cancer cells. We further discovered that reactive oxygen species accumulation activates ERK signaling, promoting cancer cell proliferation. Pro-tumorigenic macrophages exhibit fragmented mitochondrial networks, leading to higher rates of mitochondrial transfer to cancer cells. Finally, we observe that macrophage mitochondrial transfer promotes tumor cell proliferation in vivo. Collectively these results indicate that transferred macrophage mitochondria activate downstream signaling pathways in a ROS-dependent manner in cancer cells, and provide a model of how sustained behavioral reprogramming can be mediated by a relatively small amount of transferred mitochondria in vitro and in vivo. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
266. Recessive pathogenic variants in MCAT cause combined oxidative phosphorylation deficiency.
- Author
-
Webb, Bryn D., Nowinski, Sara M., Solmonson, Ashley, Ganesh, Jaya, Rodenburg, Richard J., Leandro, Joao, Evans, Anthony, Vu, Hieu S., Naidich, Thomas P., Gelb, Bruce D., DeBerardinis, Ralph J., Rutter, Jared, and Houten, Sander M.
- Subjects
- *
ACYL carrier protein , *OXIDATIVE phosphorylation , *FAILURE to thrive syndrome , *CARRIER proteins , *ELECTRON transport , *FIBROBLASTS - Abstract
Malonyl-CoA-acyl carrier protein transacylase (MCAT) is an enzyme involved in mitochondrial fatty acid synthesis (mtFAS) and catalyzes the transfer of the malonyl moiety of malonyl-CoA to the mitochondrial acyl carrier protein (ACP). Previously, we showed that loss-of-function of mtFAS genes, including Mcat, is associated with severe loss of electron transport chain (ETC) complexes in mouse immortalized skeletal myoblasts (Nowinski et al., 2020). Here, we report a proband presenting with hypotonia, failure to thrive, nystagmus, and abnormal brain MRI findings. Using whole exome sequencing, we identified biallelic variants in MCAT. Protein levels for NDUFB8 and COXII, subunits of complex I and IV respectively, were markedly reduced in lymphoblasts and fibroblasts, as well as SDHB for complex II in fibroblasts. ETC enzyme activities were decreased in parallel. Re-expression of wild-type MCAT rescued the phenotype in patient fibroblasts. This is the first report of a patient with MCAT pathogenic variants and combined oxidative phosphorylation deficiency. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
267. Collateral deletion of the mitochondrial AAA+ ATPase ATAD1 sensitizes cancer cells to proteasome dysfunction.
- Author
-
Winter, Jacob M., Fresenius, Heidi L., Cunningham, Corey N., Peng Wei, Keys, Heather R., Berg, Jordan, Bott, Alex, Yadav, Tarun, Ryan, Jeremy, Sirohi, Deepika, Tripp, Sheryl R., Barta, Paige, Agarwal, Neeraj, Letai, Anthony, Sabatini, David M., Wohlever, Matthew L., and Rutter, Jared
- Subjects
- *
TUMOR suppressor genes , *CANCER genes , *ADENOSINE triphosphatase , *MITOCHONDRIAL proteins , *MITOCHONDRIA , *CANCER cells , *HOMEOSTASIS - Abstract
The tumor suppressor gene PTEN is the second most commonly deleted gene in cancer. Such deletions often include portions of the chromosome 10q23 locus beyond the bounds of PTEN itself, which frequently disrupts adjacent genes. Coincidental loss of PTEN-adjacent genes might impose vulnerabilities that could either affect patient outcome basally or be exploited therapeutically. Here, we describe how the loss of ATAD1, which is adjacent to and frequently co-deleted with PTEN, predisposes cancer cells to apoptosis triggered by proteasome dysfunction and correlates with improved survival in cancer patients. ATAD1 directly and specifically extracts the pro-apoptotic protein BIM from mitochondria to inactivate it. Cultured cells and mouse xenografts lacking ATAD1 are hypersensitive to clinically used proteasome inhibitors, which activate BIM and trigger apoptosis. This work furthers our understanding of mitochondrial protein homeostasis and could lead to new therapeutic options for the hundreds of thousands of cancer patients who have tumors with chromosome 10q23 deletion. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
268. Intramuscular administration of glyoxylate rescues swine from lethal cyanide poisoning and ameliorates the biochemical sequalae of cyanide intoxication.
- Author
-
Bebarta, Vik S, Shi, Xu, Zheng, Shunning, Hendry-Hofer, Tara B, Severance, Carter C, Behymer, Matthew M, Boss, Gerry R, Mahon, Sari, Brenner, Matthew, Knipp, Gregory T, Davisson, Vincent Jo, Peterson, Randall T, MacRae, Calum A, Rutter, Jared, Gerszten, Robert E, and Nath, Anjali K
- Subjects
- *
CYANIDE poisoning , *POTASSIUM cyanide , *KREBS cycle , *POISONS , *SWINE , *RESPIRATION , *PULSE oximeters - Abstract
Cyanide—a fast-acting poison—is easy to obtain given its widespread use in manufacturing industries. It is a high-threat chemical agent that poses a risk of occupational exposure in addition to being a terrorist agent. FDA-approved cyanide antidotes must be given intravenously, which is not practical in a mass casualty setting due to the time and skill required to obtain intravenous access. Glyoxylate is an endogenous metabolite that binds cyanide and reverses cyanide-induced redox imbalances independent of chelation. Efficacy and biochemical mechanistic studies in an FDA-approved preclinical animal model have not been reported. Therefore, in a swine model of cyanide poisoning, we evaluated the efficacy of intramuscular glyoxylate on clinical, metabolic, and biochemical endpoints. Animals were instrumented for continuous hemodynamic monitoring and infused with potassium cyanide. Following cyanide-induced apnea, saline control or glyoxylate was administered intramuscularly. Throughout the study, serial blood samples were collected for pharmacokinetic, metabolite, and biochemical studies, in addition, vital signs, hemodynamic parameters, and laboratory values were measured. Survival in glyoxylate-treated animals was 83% compared with 12% in saline-treated control animals (p < .01). Glyoxylate treatment improved physiological parameters including pulse oximetry, arterial oxygenation, respiration, and pH. In addition, levels of citric acid cycle metabolites returned to baseline levels by the end of the study. Moreover, glyoxylate exerted distinct effects on redox balance as compared with a cyanide-chelating countermeasure. In our preclinical swine model of lethal cyanide poisoning, intramuscular administration of the endogenous metabolite glyoxylate improved survival and clinical outcomes, and ameliorated the biochemical effects of cyanide. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
269. Science SignalingPodcast: 31 January 2012
- Author
-
Cardon, Caleb M., Rutter, Jared, and VanHook, Annalisa M.
- Published
- 2012
- Full Text
- View/download PDF
270. Glyoxylate protects against cyanide toxicity through metabolic modulation.
- Author
-
Nielson, Jason R., Nath, Anjali K., Doane, Kim P., Shi, Xu, Lee, Jangwoen, Tippetts, Emily G., Saha, Kusumika, Morningstar, Jordan, Hicks, Kevin G., Chan, Adriano, Zhao, Yanbin, Kelly, Amy, Hendry-Hofer, Tara B., Witeof, Alyssa, Sips, Patrick Y., Mahon, Sari, Bebarta, Vikhyat S., Davisson, Vincent Jo, Boss, Gerry R., and Rutter, Jared
- Subjects
- *
CYANIDES , *CYTOCHROME oxidase , *POISONS , *LACTATE dehydrogenase , *SMALL molecules , *MONOCARBOXYLATE transporters - Abstract
Although cyanide's biological effects are pleiotropic, its most obvious effects are as a metabolic poison. Cyanide potently inhibits cytochrome c oxidase and potentially other metabolic enzymes, thereby unleashing a cascade of metabolic perturbations that are believed to cause lethality. From systematic screens of human metabolites using a zebrafish model of cyanide toxicity, we have identified the TCA-derived small molecule glyoxylate as a potential cyanide countermeasure. Following cyanide exposure, treatment with glyoxylate in both mammalian and non-mammalian animal models confers resistance to cyanide toxicity with greater efficacy and faster kinetics than known cyanide scavengers. Glyoxylate-mediated cyanide resistance is accompanied by rapid pyruvate consumption without an accompanying increase in lactate concentration. Lactate dehydrogenase is required for this effect which distinguishes the mechanism of glyoxylate rescue as distinct from countermeasures based solely on chemical cyanide scavenging. Our metabolic data together support the hypothesis that glyoxylate confers survival at least in part by reversing the cyanide-induced redox imbalances in the cytosol and mitochondria. The data presented herein represent the identification of a potential cyanide countermeasure operating through a novel mechanism of metabolic modulation. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
271. Paradoxical neuronal hyperexcitability in a mouse model of mitochondrial pyruvate import deficiency.
- Author
-
De La Rossa, Andres, Laporte, Marine H., Astori, Simone, Marissal, Thomas, Montessuit, Sylvie, Sheshadri, Preethi, Ramos-Fernández, Eva, Mendez, Pablo, Khani, Abbas, Quairiaux, Charles, Taylor, Eric B., Rutter, Jared, Manuel Nunes, José, Carleton, Alan, Duchen, Michael R., Sandi, Carmen, and Martinou, Jean-Claude
- Subjects
- *
LABORATORY mice , *PYRUVATES , *ANIMAL disease models , *OXIDATIVE phosphorylation , *MITOCHONDRIAL membranes , *CALCIUM channels , *POTASSIUM channels - Abstract
Neuronal excitation imposes a high demand of ATP in neurons. Most of the ATP derives primarily from pyruvate-mediated oxidative phosphorylation, a process that relies on import of pyruvate into mitochondria occuring exclusively via the mitochondrial pyruvate carrier (MPC). To investigate whether deficient oxidative phosphorylation impacts neuron excitability, we generated a mouse strain carrying a conditional deletion of MPC1, an essential subunit of the MPC, specifically in adult glutamatergic neurons. We found that, despite decreased levels of oxidative phosphorylation and decreased mitochondrial membrane potential in these excitatory neurons, mice were normal at rest. Surprisingly, in response to mild inhibition of GABA mediated synaptic activity, they rapidly developed severe seizures and died, whereas under similar conditions the behavior of control mice remained unchanged. We report that neurons with a deficient MPC were intrinsically hyperexcitable as a consequence of impaired calcium homeostasis, which reduced M-type potassium channel activity. Provision of ketone bodies restored energy status, calcium homeostasis and M-channel activity and attenuated seizures in animals fed a ketogenic diet. Our results provide an explanation for the seizures that frequently accompany a large number of neuropathologies, including cerebral ischemia and diverse mitochondriopathies, in which neurons experience an energy deficit. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
272. Protective mitochondrial fission induced by stress-responsive protein GJA1-20k.
- Author
-
Shimura, Daisuke, Nuebel, Esther, Baum, Rachel, Valdez, Steven E., Shaohua Xiao, Warren, Junco S., Palatinus, Joseph A., TingTing Hong, Rutter, Jared, and Shaw, Robin M.
- Subjects
- *
REPERFUSION injury , *MITOCHONDRIA , *REACTIVE oxygen species , *ISCHEMIC preconditioning , *HEART , *PROTEINS , *CONNEXIN 43 , *MYOCARDIAL infarction - Abstract
The Connexin43 gap junction gene GJA1 has one coding exon, but its mRNA undergoes internal translation to generate N-terminal truncated isoforms of Connexin43 with the predominant isoform being only 20 kDa in size (GJA1-20k). Endogenous GJA1-20k protein is not membrane bound and has been found to increase in response to ischemic stress, localize to mitochondria, and mimic ischemic preconditioning protection in the heart. However, it is not known how GJA1-20k benefits mitochondria to provide this protection. Here, using human cells and mice, we identify that GJA1-20k polymerizes actin around mitochondria which induces focal constriction sites. Mitochondrial fission events occur within about 45 s of GJA1-20k recruitment of actin. Interestingly, GJA1-20k mediated fission is independent of canonical Dynamin-Related Protein 1 (DRP1). We find that GJA1-20k-induced smaller mitochondria have decreased reactive oxygen species (ROS) generation and, in hearts, provide potent protection against ischemia-reperfusion injury. The results indicate that stress responsive internally translated GJA1-20k stabilizes polymerized actin filaments to stimulate non-canonical mitochondrial fission which limits ischemic-reperfusion induced myocardial infarction. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
273. Identification of small molecule allosteric modulators of 5,10-methylenetetrahydrofolate reductase (MTHFR) by targeting its unique regulatory domain.
- Author
-
Bezerra, Gustavo A., Holenstein, Alexander, Foster, William R., Xie, Bing, Hicks, Kevin G., Bürer, Céline, Lutz, Seraina, Mukherjee, Ayan, Sarkar, Dipika, Bhattacharya, Debomita, Rutter, Jared, Talukdar, Arindam, Brown, Peter J., Luo, Minkui, Shi, Lei, Froese, D. Sean, and Yue, Wyatt W.
- Subjects
- *
SMALL molecules , *PROTEIN arginine methyltransferases , *SURFACE plasmon resonance , *BINDING sites , *BINDING site assay - Abstract
The folate and methionine cycles, constituting one-carbon metabolism, are critical pathways for cell survival. Intersecting these two cycles, 5,10-methylenetetrahydrofolate reductase (MTHFR) directs one-carbon units from the folate to methionine cycle, to be exclusively used for methionine and S -adenosylmethionine (AdoMet) synthesis. MTHFR deficiency and upregulation result in diverse disease states, rendering it an attractive drug target. The activity of MTHFR is inhibited by the binding of AdoMet to an allosteric regulatory domain distal to the enzyme's active site, which we have previously identified to constitute a novel fold with a druggable pocket. Here, we screened 162 AdoMet mimetics using differential scanning fluorimetry, and identified 4 compounds that stabilized this regulatory domain. Three compounds were sinefungin analogues, closely related to AdoMet and S -adenosylhomocysteine (AdoHcy). The strongest thermal stabilisation was provided by (S)-SKI-72, a potent inhibitor originally developed for protein arginine methyltransferase 4 (PRMT4). Using surface plasmon resonance, we confirmed that (S)-SKI-72 binds MTHFR via its allosteric domain with nanomolar affinity. Assay of MTHFR activity in the presence of (S)-SKI-72 demonstrates inhibition of purified enzyme with sub-micromolar potency and endogenous MTHFR from HEK293 cell lysate in the low micromolar range, both of which are lower than AdoMet. Nevertheless, unlike AdoMet, (S)-SKI-72 is unable to completely abolish MTHFR activity, even at very high concentrations. Combining binding assays, kinetic characterization and compound docking, this work indicates the regulatory domain of MTHFR can be targeted by small molecules and presents (S)-SKI-72 as an excellent candidate for development of MTHFR inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
274. Integrating the dynamic and energetic fields of metabolism and development.
- Author
-
Finley, Lydia, Gendron, Joshua, Miguel-Aliaga, Irene, and Rutter, Jared
- Subjects
- *
MEDICAL sciences , *BIOLOGISTS , *CELL determination , *PLANT development , *IRON metabolism , *FETAL development - Abstract
This document is an editorial that discusses the integration of the fields of metabolism and development. It highlights the role of metabolic processes in cellular energy production and the production of cellular components. The editorial emphasizes the emerging understanding that metabolites can also influence gene expression and cell behavior. The document includes research articles that explore the relationship between metabolism and development in various systems, including plants and animals. It also includes review articles that provide an overview of the nutritional control of development and the specific roles of nutrients in mammalian development. Overall, the document showcases the diverse functions of metabolism in the context of developmental biology. [Extracted from the article]
- Published
- 2023
- Full Text
- View/download PDF
275. Mitochondrial pyruvate carrier is required for optimal brown fat thermogenesis.
- Author
-
Panic, Vanja, Pearson, Stephanie, Banks, James, Tippetts, Trevor S., Velasco-Silva, Jesse N., Sanghoon Lee, Simcox, Judith, Geoghegan, Gisela, Bensard, Claire L., van Ry, Tyler, Holland, Will L., Summers, Scott A., Cox, James, Ducker, Gregory S., Rutter, Jared, and Villanueva, Claudio J.
- Subjects
- *
BROWN adipose tissue , *BODY temperature , *BODY temperature regulation , *ALTERNATIVE fuels , *COLD adaptation , *OXIDATION of glucose - Abstract
Brown adipose tissue (BAT) is composed of thermogenic cells that convert chemical energy into heat to maintain a constant body temperature and counteract metabolic disease. The metabolic adaptations required for thermogenesis are not fully understood. Here, we explore how steady state levels of metabolic intermediates are altered in brown adipose tissue in response to cold exposure. Transcriptome and metabolome analysis revealed changes in pathways involved in amino acid, glucose, and TCA cycle metabolism. Using isotopic labeling experiments, we found that activated brown adipocytes increased labeling of pyruvate and TCA cycle intermediates from U13C-glucose. Although glucose oxidation has been implicated as being essential for thermogenesis, its requirement for efficient thermogenesis has not been directly tested. We show that mitochondrial pyruvate uptake is essential for optimal thermogenesis, as conditional deletion of Mpc1 in brown adipocytes leads to impaired cold adaptation. Isotopic labeling experiments using U13C-glucose showed that loss of MPC1 led to impaired labeling of TCA cycle intermediates. Loss of MPC1 in BAT increased 3-hydroxybutyrate levels in blood and BAT in response to the cold, suggesting that ketogenesis provides an alternative fuel source to compensate. Collectively, these studies highlight that complete glucose oxidation is essential for optimal brown fat thermogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
276. Mitochondrial fatty acid synthesis coordinates oxidative metabolism in mammalian mitochondria.
- Author
-
Nowinski, Sara M., Solmonson, Ashley, Rusin, Scott F., Maschek, J. Alan, Bensard, Claire L., Fogarty, Sarah, Mi-Young Jeong, Lettlova, Sandra, Berg, Jordan A., Morgan, Jeffrey T., Yeyun Ouyang, Naylor, Bradley C., Paulo, Joao A., Katsuhiko Funai, Cox, James E., Gygi, Steven P., Winge, Dennis R., DeBerardinis, Ralph J., and Rutter, Jared
- Subjects
- *
FATTY acids , *FATTY acid oxidation , *MITOCHONDRIA , *ELECTRON transport , *METABOLISM - Abstract
Cells harbor two systems for fatty acid synthesis, one in the cytoplasm (catalyzed by fatty acid synthase, FASN) and one in the mitochondria (mtFAS). In contrast to FASN, mtFAS is poorly characterized, especially in higher eukaryotes, with the major product(s), metabolic roles, and cellular function(s) being essentially unknown. Here we show that hypomorphic mtFAS mutant mouse skeletal myoblast cell lines display a severe loss of electron transport chain (ETC) complexes and exhibit compensatory metabolic activities including reductive carboxylation. This effect on ETC complexes appears to be independent of protein lipoylation, the best characterized function of mtFAS, as mutants lacking lipoylation have an intact ETC. Finally, mtFAS impairment blocks the differentiation of skeletal myoblasts in vitro. Together, these data suggest that ETC activity in mammals is profoundly controlled by mtFAS function, thereby connecting anabolic fatty acid synthesis with the oxidation of carbon fuels. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
277. The Role of Nonglycolytic Glucose Metabolism in Myocardial Recovery Upon Mechanical Unloading and Circulatory Support in Chronic Heart Failure.
- Author
-
Badolia, Rachit, Ramadurai, Dinesh K.A., Abel, E. Dale, Ferrin, Peter, Taleb, Iosif, Shankar, Thirupura S., Krokidi, Aspasia Thodou, Navankasattusas, Sutip, McKellar, Stephen H., Yin, Michael, Kfoury, Abdallah G., Wever-Pinzon, Omar, Fang, James C., Selzman, Craig H., Chaudhuri, Dipayan, Rutter, Jared, Drakos, Stavros G., and Thodou Krokidi, Aspasia
- Subjects
- *
HEART metabolism , *GLUCOSE metabolism , *NICOTINAMIDE adenine dinucleotide phosphate , *REACTIVE oxygen species , *PENTOSE phosphate pathway , *BIOCHEMISTRY , *ENERGY metabolism , *RESEARCH , *RESEARCH methodology , *METABOLISM , *HEART assist devices , *MEDICAL cooperation , *EVALUATION research , *HEART ventricles , *COMPARATIVE studies , *RESEARCH funding , *STROKE volume (Cardiac output) , *HEART failure , *GLYCOLYSIS , *COMORBIDITY , *OXIDATION-reduction reaction - Abstract
Background: Significant improvements in myocardial structure and function have been reported in some patients with advanced heart failure (termed responders [R]) following left ventricular assist device (LVAD)-induced mechanical unloading. This therapeutic strategy may alter myocardial energy metabolism in a manner that reverses the deleterious metabolic adaptations of the failing heart. Specifically, our previous work demonstrated a post-LVAD dissociation of glycolysis and oxidative-phosphorylation characterized by induction of glycolysis without subsequent increase in pyruvate oxidation through the tricarboxylic acid cycle. The underlying mechanisms responsible for this dissociation are not well understood. We hypothesized that the accumulated glycolytic intermediates are channeled into cardioprotective and repair pathways, such as the pentose-phosphate pathway and 1-carbon metabolism, which may mediate myocardial recovery in R.Methods: We prospectively obtained paired left ventricular apical myocardial tissue from nonfailing donor hearts as well as R and nonresponders at LVAD implantation (pre-LVAD) and transplantation (post-LVAD). We conducted protein expression and metabolite profiling and evaluated mitochondrial structure using electron microscopy.Results: Western blot analysis shows significant increase in rate-limiting enzymes of pentose-phosphate pathway and 1-carbon metabolism in post-LVAD R (post-R) as compared with post-LVAD nonresponders (post-NR). The metabolite levels of these enzyme substrates, such as sedoheptulose-6-phosphate (pentose phosphate pathway) and serine and glycine (1-carbon metabolism) were also decreased in Post-R. Furthermore, post-R had significantly higher reduced nicotinamide adenine dinucleotide phosphate levels, reduced reactive oxygen species levels, improved mitochondrial density, and enhanced glycosylation of the extracellular matrix protein, α-dystroglycan, all consistent with enhanced pentose-phosphate pathway and 1-carbon metabolism that correlated with the observed myocardial recovery.Conclusions: The recovering heart appears to direct glycolytic metabolites into pentose-phosphate pathway and 1-carbon metabolism, which could contribute to cardioprotection by generating reduced nicotinamide adenine dinucleotide phosphate to enhance biosynthesis and by reducing oxidative stress. These findings provide further insights into mechanisms responsible for the beneficial effect of glycolysis induction during the recovery of failing human hearts after mechanical unloading. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
278. T Cell-Expressed microRNA-155 Reduces Lifespan in a Mouse Model of Age-Related Chronic Inflammation.
- Author
-
Ekiz, H. Atakan, Ramstead, Andrew G., Soh-Hyun Lee, Nelson, Morgan C., Bauer, Kaylyn M., Wallace, Jared A., Ruozhen Hu, Round, June L., Rutter, Jared, Drummond, Micah J., Rao, Dinesh S., and O'Connell, Ryan M.
- Subjects
- *
T helper cells , *LABORATORY mice , *T cells , *ANIMAL disease models , *CELL populations - Abstract
Aging-related chronic inflammation is a risk factor for many human disorders through incompletely understood mechanisms. Aged mice deficient in microRNA (miRNA/miR)-146a succumb to life-shortening chronic inflammation. In this study, we report that miR-155 in T cells contributes to shortened lifespan of miR-146a-/- mice. Using single-cell RNA sequencing and flow cytometry, we found that miR-155 promotes the activation of effector T cell populations, including T follicular helper cells, and increases germinal center B cells and autoantibodies in mice aged over 15 months. Mechanistically, aerobic glycolysis genes are elevated in T cells during aging, and upon deletion of miR-146a, in a T cell miR-155-dependent manner. Finally, skewing T cell metabolism toward aerobic glycolysis by deleting mitochondrial pyruvate carrier recapitulates age-dependent T cell phenotypes observed in miR-146a-/- mice, revealing the sufficiency of metabolic reprogramming to influence immune cell functions during aging. Altogether, these data indicate that T cell-specific miRNAs play pivotal roles in regulating lifespan through their influences on inflammaging. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
279. XPRESSyourself: Enhancing, standardizing, and automating ribosome profiling computational analyses yields improved insight into data.
- Author
-
Berg, Jordan A., Belyeu, Jonathan R., Morgan, Jeffrey T., Ouyang, Yeyun, Bott, Alex J., Quinlan, Aaron R., Gertz, Jason, and Rutter, Jared
- Subjects
- *
RIBOSOMES , *NUCLEIC acids , *DATA analysis , *MACHINE translating , *BEST practices , *DATA - Abstract
Ribosome profiling, an application of nucleic acid sequencing for monitoring ribosome activity, has revolutionized our understanding of protein translation dynamics. This technique has been available for a decade, yet the current state and standardization of publicly available computational tools for these data is bleak. We introduce XPRESSyourself, an analytical toolkit that eliminates barriers and bottlenecks associated with this specialized data type by filling gaps in the computational toolset for both experts and non-experts of ribosome profiling. XPRESSyourself automates and standardizes analysis procedures, decreasing time-to-discovery and increasing reproducibility. This toolkit acts as a reference implementation of current best practices in ribosome profiling analysis. We demonstrate this toolkit's performance on publicly available ribosome profiling data by rapidly identifying hypothetical mechanisms related to neurodegenerative phenotypes and neuroprotective mechanisms of the small-molecule ISRIB during acute cellular stress. XPRESSyourself brings robust, rapid analysis of ribosome-profiling data to a broad and ever-expanding audience and will lead to more reproducible and accessible measurements of translation regulation. XPRESSyourself software is perpetually open-source under the GPL-3.0 license and is hosted at https://github.com/XPRESSyourself, where users can access additional documentation and report software issues. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
280. Activation of PASK by mTORC1 is required for the onset of the terminal differentiation program.
- Author
-
Kikani, Chintan K., Xiaoying Wu, Fogarty, Sarah, Seong Anthony Woo Kang, Dephoure, Noah, Gygi, Steven P., Sabatini, David M., and Rutter, Jared
- Subjects
- *
STEM cells , *MYOGENIN , *HYPERTROPHY , *PHOSPHORYLATION , *MYOBLASTS - Abstract
During skeletal muscle regeneration, muscle stem cells (MuSCs) respond to multiple signaling inputs that converge onto mammalian target of rapamycin complex 1 (mTORC1) signaling pathways. mTOR function is essential for establishment of the differentiationcommitted progenitors (early stage of differentiation, marked by the induction of myogenin expression), myotube fusion, and, ultimately, hypertrophy (later stage of differentiation). While a major mTORC1 substrate, p70S6K, is required for myotube fusion and hypertrophy, an mTORC1 effector for the induction of myogenin expression remains unclear. Here, we identified Per-Arnt-Sim domain kinase (PASK) as a downstream phosphorylation target ofmTORC1 in MuSCs during differentiation. We have recently shown that the PASK phosphorylates Wdr5 to stimulate MuSC differentiation by epigenetically activating the myogenin promoter. We show that phosphorylation of PASK by mTORC1 is required for the activation of myogenin transcription, exit from self-renewal, and induction of the myogenesis program. Our studies reveal that mTORC1-PASK signaling is required for the rise of myogenin-positive committed myoblasts (early stage of myogenesis), whereas mTORC1-S6K signaling is required for myoblast fusion (later stage of myogenesis). Thus, our discoveries allow molecular dissection of mTOR functions during different stages of the myogenesis program driven by two different substrates. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
281. Structure of human Fe-S assembly subcomplex reveals unexpected cysteine desulfurase architecture and acyl-ACP-ISD11 interactions.
- Author
-
Cory, Seth A., Van Vranken, Jonathan G., Brignole, Edward J., Patra, Shachin, Winge, Dennis R., Drennan, Catherine L., Rutter, Jared, and Barondeau, David P.
- Subjects
- *
CYSTEINE desulfurase , *LYASES , *SULFOTRANSFERASES , *CARRIER proteins , *PROTEIN binding - Abstract
In eukaryotes, sulfur is mobilized for incorporation into multiple biosynthetic pathways by a cysteine desulfurase complex that consists of a catalytic subunit (NFS1), LYR protein (ISD11) and acyl carrier protein (ACP). This NFS1-ISD11-ACP (SDA) complex forms the core of the iron-sulfur (Fe-S) assembly complex and associates with assembly proteins ISCU2, frataxin (FXN) and ferredoxin to synthesize Fe-S clusters. Here we present crystallographic and electron microscopic structures of the SDA complex coupled to enzyme kinetic and cell-based studies to provide structure-function properties of a mitochondrial cysteine desulfurase. Unlike prokaryotic cysteine desulfurases, the SDA structure adopts an unexpected architecture in which a pair of ISD11 subunits form the dimeric core of the SDA complex, which clarifies the critical role of ISD11 in eukaryotic assemblies. The different quaternary structure results in an incompletely formed substrate channel and solvent-exposed pyridoxal 5'-phosphate cofactor and provides a rationale for the allosteric activator function of FXN in eukaryotic systems. The structure also reveals the 4'-phosphopantetheine-conjugated acyl-group of ACP occupies the hydrophobic core of ISD11, explaining the basis of ACP stabilization. The unexpected architecture for the SDA complex provides a framework for understanding interactions with acceptor proteins for sulfur-containing biosynthetic pathways, elucidating mechanistic details of eukaryotic Fe-S cluster biosynthesis and clarifying how defects in Fe-S cluster assembly lead to diseases such as Friedreich's ataxia. Moreover, our results support a lock-and-key model in which LYR proteins associate with acyl-ACP as a mechanism for fatty acid biosynthesis to coordinate the expression, Fe-S cofactor maturation and activity of the respiratory complexes. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
282. Per-Arnt-Sim Kinase Regulates Pancreatic Duodenal Homeobox-1 Protein Stability via Phosphorylation of Glycogen Synthase Kinase 3β in Pancreatic β-Cells.
- Author
-
Semache, Meriem, Zarrouki, Bader, Fontés, Ghislaine, Fogarty, Sarah, Kikani, Chintan, Chawki, Mohammad B., Rutter, Jared, and Poitout, Vincent
- Subjects
- *
GLUCOSE , *TRANSCRIPTION factors , *DUODENUM , *PHOSPHORYLATION , *SERINE - Abstract
In pancreatic β-cells, glucose induces the binding of the transcription factor pancreatic duodenal homeobox-1 (PDX-1) to the insulin gene promoter to activate insulin gene transcription. At low glucose levels, glycogen synthase kinase 3β (GSK3β) is known to phosphorylate PDX-1 on C-terminal serine residues, which triggers PDX-1 proteasomal degradation. We previously showed that the serine/threonine Per-Arnt-Sim domain-containing kinase (PASK) regulates insulin gene transcription via PDX-1. However, the mechanisms underlying this regulation are unknown. In this study, we aimed to identify the role of PASK in the regulation of PDX-1 phosphorylation, protein expression, and stability in insulin-secreting cells and isolated rodent islets of Langerhans. We observed that glucose induces a decrease in overall PDX-1 serine phosphorylation and that over- expression of WT PASK mimics this effect. In vitro, PASK directly phosphorylates GSK3β on its inactivating phosphorylation site Ser9 . Overexpression of a kinase-dead (KD), dominant negative version of PASK blocks glucose-induced Ser9 phosphorylation of GSK3β. Accordingly, GSK3β Ser9 phosphorylation is reduced in islets from pask-null mice. Overexpression of WT PASK or KDGSK3 protects PDX-1 from degradation and results in increased PDX-1 protein abundance. Conversely, over- expression of KD PASK blocks glucose-induction of PDX-1 protein. We conclude that PASK phosphorylates and inactivates GSK3β, thereby preventing PDX-1 serine phosphorylation and alleviating GSK3β-mediated PDX-1 protein degradation in pancreatic-cells. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
283. A Mitochondrial Pyruvate Carrier Required for Pyruvate Uptake in Yeast, Drosophila, and Humans.
- Author
-
Bricker, Daniel K., Taylor, Eric B., Schell, John C., Orsak, Thomas, Boutron, Audrey, Yu-Chan Chen, Cox, James E., Cardon, Caleb M., Van Vranken, Jonathan G., Dephoure, Noah, Redin, Claire, Boudina, Sihem, Gygi, Steven P., Brivet, Michèle, Thummel, Carl S., and Rutter, Jared
- Subjects
- *
PYRUVATES , *MITOCHONDRIAL proteins , *BIOLOGICAL transport , *CARRIER proteins , *MITOCHONDRIAL membranes , *CARBON metabolism , *FUNGAL proteins , *HUMAN proteins , *DROSOPHILA proteins - Abstract
Pyruvate constitutes a critical branch point in cellular carbon metabolism. We have identified two proteins, Mpc1 and Mpc2, as essential for mitochondrial pyruvate transport in yeast, Drosophila, and humans. Mpc1 and Mpc2 associate to form an ~150-kitodalton complex in the inner mitochondrial membrane. Yeast and Drosophila mutants lacking MPC1 display impaired pyruvate metabolism, with an accumulation of upstream metabolites and a depletion of tricarboxylic acid cycle intermediates. Loss of yeast Mpc1 results in defective mitochondrial pyruvate uptake, and silencing of MPC1 or MPC2 in mammalian cells impairs pyruvate oxidation. A point mutation in MPC1 provides resistance to a known inhibitor of the mitochondrial pyruvate carrier. Human genetic studies of three families with children suffering from lactic acidosis and hyperpyruvatemia revealed a causal locus that mapped to MPC1, changing single amino acids that are conserved throughout eukaryotes. These data demonstrate that Mpc1 and Mpc2 form an essential part of the mitochondrial pyruvate carrier. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
284. SDH5, a Gene Required for Flavination of Succinate Dehydrogenase, Is Mutated in Paraganglioma.
- Author
-
Huai-Xiang Hao, Khalimonchuk, Oleh, Schraders, Margit, Dephoure, Noah, Bayley, Jean-Pierre, Kunst, Henricus, Devilee, Peter, Cremers, Cor W. R. J., Schiffman, Joshua D., Bentz, Brandon G., Gygi, Steven P., Winge, Dennis R., Kremer, Hannie, and Rutter, Jared
- Subjects
- *
SUCCINATE dehydrogenase , *PARAGANGLIOMA , *GENETIC mutation , *MITOCHONDRIA , *PROTEIN research , *YEAST research , *KREBS cycle , *PROTEOMICS - Abstract
Mammalian mitochondria contain about 1100 proteins, nearly 300 of which are uncharacterized. Given the well-established role of mitochondrial defects in human disease, functional characterization of these proteins may shed new tight on disease mechanisms. Starting with yeast as a model system, we investigated an uncharacterized but highly conserved mitochondrial protein (named here Sdh5). Both yeast and human Sdh5 interact with the catalytic subunit of the succinate dehydrogenase (SDH) complex, a component of both the electron transport chain and the tricarboxylic acid cycle. Sdh5 is required for SDH-dependent respiration and for Sdh1 flavination (incorporation of the flavin adenine dinucleotide cofactor). Germline loss-of-function mutations in the human SDH5 gene, located on chromosome 11q13.1, segregate with disease in a family with hereditary paraganglioma, a neuroendocrine tumor previously linked to mutations in genes encoding SDH subunits. Thus, a mitochondrial proteomics analysis in yeast has led to the discovery of a human tumor susceptibility gene. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
285. Efficient gene targeting in Drosophila by direct embryo injection with zinc-finger nucleases.
- Author
-
Beumer, Kelly J., Trautman, Jonathan K., Bozas, Ana, Ji-Long Liu, Rutter, Jared, GalI, Joseph G., and Carroll, Dana
- Subjects
- *
GENE targeting , *DROSOPHILA melanogaster , *NUCLEASES , *MUTAGENESIS , *DNA repair , *ZINC-finger proteins - Abstract
We report very high gene targeting frequencies in Drosophila by direct embryo injection of mRNA5 encoding specific zinc-finger nucleases (ZFNs). Both local mutagenesis via nonhomologous end joining (NHEJ) and targeted gene replacement via homologous recombination (HR) have been achieved in up to 10% of all targets at a given locus. In embryos that are wild type for DNA repair, the products are dominated by NHEJ mutations. In recipients deficient in the NHEJ component, DNA ligase IV, the majority of products arise by HR with a coinjected donor DNA. with no loss of overall efficiency in target modification. We describe the application of the ZFN injection procedure to mutagenesis by NHEJ of 2 new genes in Drosophila melanogaster: coil and pask. Pairs of novel ZFN5 designed for targets within those genes led to the production of null mutations at each locus. The injection procedure is much more rapid than earlier approaches and makes possible the generation and recovery of targeted gene alterations at essentially any locus within 2 fly generations. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
286. Yeast PAS kinase coordinates glucose partitioning in response to metabolic and cell integrity signaling.
- Author
-
Grose, Julianne H., Smith, Tammy L., Sabic, Hana, and Rutter, Jared
- Subjects
- *
PROTEIN kinases , *PHOSPHOTRANSFERASES , *PHYSIOLOGICAL control systems , *BIOMOLECULES , *CHEMICAL reactions , *BIOCHEMISTRY - Abstract
PAS kinase is an evolutionarily conserved serine/threonine protein kinase. Mammalian PAS kinase is activated under nutrient replete conditions and is important for controlling metabolic rate and energy homeostasis. In yeast, PAS kinase acts to increase the synthesis of structural carbohydrate at the expense of storage carbohydrates through phosphorylation of the enzyme UDP-glucose pyrophosphorylase. We have identified two pathways that activate yeast PAS kinase; one is responsive to nutrient conditions while the other is responsive to cell integrity stress. These pathways differentially activate the two PAS kinase proteins in Saccharomyces cerevisiae, Psk1 and Psk2, with Psk1 alone responding to activation by nonfermentative carbon sources. We demonstrate that, in addition to transcriptional effects, both of these pathways post-translationally activate PAS kinase via its regulatory N-terminus. As a whole, this system acts to coordinate glucose partitioning with alterations in demand due to changes in environmental and nutrient conditions. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
287. Control of mammalian glycogen synthase by PAS kinase.
- Author
-
Wilson, Wayne A., Skurat, Alexander V., Probst, Brandon, De Paoli-Roach, Anna, Roach, Peter J., and Rutter, Jared
- Subjects
- *
GLUCANS , *PHOSPHORYLATION , *PHYSIOLOGICAL control systems , *SACCHAROMYCES cerevisiae , *BIOCHEMISTRY , *POLYALUMINUM sulfate , *ENZYMES - Abstract
The regulation of glycogen metabolism is critical for the maintenance of glucose and energy homeostasis in mammals. Glycogen synthase, the enzyme responsible for glycogen production, is regulated by multisite phosphorylation in yeast and mammals. We have previously identified PAS kinase as a physiological regulator of glycogen synthase in Saccharomyces cerevisiae. We provide evidence here that PAS kinase is an important regulator of mammalian glycogen synthase. Glycogen synthase is efficiently phosphorylated by PAS kinase in vitro at Ser-640, a known regulatory phosphosite. Efficient phosphorylation requires a region of PAS kinase outside the catalytic domain. This region appears to mediate a direct interaction between glycogen synthase and PAS kinase, thereby targeting kinase activity to this substrate specifically. This interaction is regulated by the PAS kinase PAS domain, raising the possibility that this interaction (and phosphorylation event) is modulated by the cellular metabolic state. This mode of regulation provides a mechanism for metabolic status to impinge directly on the cellular decision of whether to store or use available energy. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
288. Impaired Cued and Contextual Memory in NPAS2-Deficient Mice.
- Author
-
Garcia, Joseph A., Zhang, Di, Estill, Sandi Jo, Michnoff, Carolyn, Rutter, Jared, Reick, Martin, Scott, Kristin, Diaz-Arrastia, Ramon, and McKnight, Steven L.
- Subjects
- *
MEMORY research , *MICE , *T cells , *NEUROGENETICS , *BETA-galactosidase , *PHYSIOLOGY - Abstract
Provides information on a study which investigated the role of neuronal PAS domain protein 2 (NPAS2) in long-term memory arm of the cued and contextual fear task of mice. Definition of NPAS2; Examination on the origin of T-cell independent immunoglobulin A-producing B cells; Description on the neuroanatomical expression pattern of beta-galactosidase activity in NPAS2-lacZ mice.
- Published
- 2000
- Full Text
- View/download PDF
289. Evaluation of EGFR and COX pathway inhibition in human colon organoids of serrated polyposis and other hereditary cancer syndromes.
- Author
-
Kanth P, Hazel MW, Schell JC, Rutter J, Yao R, Mills AP, and Delker DA
- Subjects
- Humans, Sulindac pharmacology, Colorectal Neoplasms, Hereditary Nonpolyposis genetics, Colorectal Neoplasms, Hereditary Nonpolyposis drug therapy, Colorectal Neoplasms, Hereditary Nonpolyposis pathology, Colonic Polyps genetics, Colonic Polyps pathology, Colonic Polyps drug therapy, Female, Middle Aged, Colon pathology, Colon drug effects, Male, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins B-raf genetics, Mutation, Adult, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Signal Transduction drug effects, Neoplastic Syndromes, Hereditary genetics, Neoplastic Syndromes, Hereditary drug therapy, Neoplastic Syndromes, Hereditary pathology, Organoids drug effects, Organoids pathology, Organoids metabolism, ErbB Receptors genetics, Adenomatous Polyposis Coli genetics, Adenomatous Polyposis Coli pathology, Adenomatous Polyposis Coli drug therapy, Erlotinib Hydrochloride pharmacology, Erlotinib Hydrochloride therapeutic use
- Abstract
Serrated polyposis syndrome (SPS) presents with multiple sessile serrated lesions (SSL) in the large intestine and confers increased colorectal cancer (CRC) risk. However, the etiology of SPS is not known. SSL-derived organoids have not been previously studied but may help provide insights into SPS pathogenesis and identify novel biomarkers and chemopreventive strategies. This study examined effects of EGFR and COX pathway inhibition in organoid cultures derived from uninvolved colon and polyps of SPS patients. We also compared with organoids representing the hereditary gastrointestinal syndromes, Familial Adenomatous Polyposis (FAP) and Lynch syndrome (LS). Eighteen total organoid colon cultures were generated from uninvolved colon and polyps in SPS, FAP, LS, and non-syndromic screening colonoscopy patients. BRAF and KRAS mutation status was determined for each culture. Erlotinib (EGFR inhibitor) and sulindac (COX inhibitor) were applied individually and in combination. A 44-target gene custom mRNA panel (including WNT and COX pathway genes) and a 798-gene microRNA gene panel were used to quantitate organoid RNA expression by NanoString analysis. Erlotinib treatment significantly decreased levels of mRNAs associated with WNT and MAPK kinase signaling in organoids from uninvolved colon from all four patient categories and from all SSL and adenomatous polyps. Sulindac did not change the mRNA profile in any culture. Our findings suggest that EGFR inhibitors may contribute to the chemopreventive treatment of SSLs. These findings may also facilitate clinical trial design using these agents in SPS patients. Differentially expressed genes identified in our study (MYC, FOSL1, EGR1, IL33, LGR5 and FOXQ1) may be used to identify other new molecular targets for chemoprevention of SSLs., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
290. Direct mitochondrial import of lactate supports resilient carbohydrate oxidation.
- Author
-
Cluntun AA, Visker JR, Velasco-Silva JN, Lang MJ, Cedeño-Rosario L, Shankar TS, Hamouche R, Ling J, Kim JE, Toshniwal AG, Low HK, Cunningham CN, Carrington J, Catrow JL, Pearce Q, Jeong MY, Bott AJ, Narbona-Pérez ÁJ, Stanley CE, Li Q, Eberhardt DR, Morgan JT, Yadav T, Wells CE, Ramadurai DKA, Swiatek WI, Chaudhuri D, Rothstein JD, Muoio DM, Paulo JA, Gygi SP, Baker SA, Navankasattusas S, Cox JE, Funai K, Drakos SG, Rutter J, and Ducker GS
- Abstract
Lactate is the highest turnover circulating metabolite in mammals. While traditionally viewed as a waste product, lactate is an important energy source for many organs, but first must be oxidized to pyruvate for entry into the tricarboxylic acid cycle (TCA cycle). This reaction is thought to occur in the cytosol, with pyruvate subsequently transported into mitochondria via the mitochondrial pyruvate carrier (MPC). Using
13 C stable isotope tracing, we demonstrated that lactate is oxidized in the myocardial tissue of mice even when the MPC is genetically deleted. This MPC-independent lactate import and mitochondrial oxidation is dependent upon the monocarboxylate transporter 1 (MCT1/ Slc16a1 ). Mitochondria isolated from the myocardium without MCT1 exhibit a specific defect in mitochondrial lactate, but not pyruvate, metabolism. The import and subsequent mitochondrial oxidation of lactate by mitochondrial lactate dehydrogenase (LDH) acts as an electron shuttle, generating sufficient NADH to support respiration even when the TCA cycle is disrupted. In response to diverse cardiac insults, animals with hearts lacking MCT1 undergo rapid progression to heart failure with reduced ejection fraction. Thus, the mitochondrial import and oxidation of lactate enables carbohydrate entry into the TCA cycle to sustain cardiac energetics and maintain myocardial structure and function under stress conditions., Competing Interests: Disclosures S.G.D. serves as a consultant for Abbott Laboratories and Pfizer. S.G.D and J.R have received research support from Novartis and Merck. The remaining authors declare no competing interests or financial relationships.- Published
- 2024
- Full Text
- View/download PDF
291. The fate of pyruvate dictates cell growth by modulating cellular redox potential.
- Author
-
Toshniwal AG, Lam G, Bott AJ, Cluntun AA, Skabelund R, Nam HJ, Wisidagama DR, Thummel CS, and Rutter J
- Abstract
Pyruvate occupies a central node in carbohydrate metabolism such that how it is produced and consumed can optimize a cell for energy production or biosynthetic capacity. This has been primarily studied in proliferating cells, but observations from the post-mitotic Drosophila fat body led us to hypothesize that pyruvate fate might dictate the rapid cell growth observed in this organ during development. Indeed, we demonstrate that augmented mitochondrial pyruvate import prevented cell growth in fat body cells in vivo as well as in cultured mammalian hepatocytes and human hepatocyte-derived cells in vitro . This effect on cell size was caused by an increase in the NADH/NAD
+ ratio, which rewired metabolism toward gluconeogenesis and suppressed the biomass-supporting glycolytic pathway. Amino acid synthesis was decreased, and the resulting loss of protein synthesis prevented cell growth. Surprisingly, this all occurred in the face of activated pro-growth signaling pathways, including mTORC1, Myc, and PI3K/Akt. These observations highlight the evolutionarily conserved role of pyruvate metabolism in setting the balance between energy extraction and biomass production in specialized post-mitotic cells.- Published
- 2024
- Full Text
- View/download PDF
292. Enhancing mitochondrial pyruvate metabolism ameliorates ischemic reperfusion injury in the heart.
- Author
-
Visker JR, Cluntun AA, Velasco-Silva JN, Eberhardt DR, Cedeño-Rosario L, Shankar TS, Hamouche R, Ling J, Kwak H, Hillas JY, Aist I, Tseliou E, Navankasattusas S, Chaudhuri D, Ducker GS, Drakos SG, and Rutter J
- Subjects
- Animals, Mice, Male, Muscle Proteins metabolism, Myocardium metabolism, Myocardium pathology, Mitochondria, Heart metabolism, Lactic Acid metabolism, Disease Models, Animal, Myocardial Infarction metabolism, Mitochondria metabolism, Oxidation-Reduction, Reactive Oxygen Species metabolism, Mice, Inbred C57BL, Anion Transport Proteins, Myocardial Reperfusion Injury metabolism, Monocarboxylic Acid Transporters metabolism, Pyruvic Acid metabolism, Myocytes, Cardiac metabolism, Mitochondrial Membrane Transport Proteins metabolism
- Abstract
The clinical therapy for treating acute myocardial infarction is primary percutaneous coronary intervention (PPCI). PPCI is effective at reperfusing the heart; however, the rapid reintroduction of blood can cause ischemia-reperfusion (I/R). Reperfusion injury is responsible for up to half of the total myocardial damage, but there are no pharmacological interventions to reduce I/R. We previously demonstrated that inhibiting monocarboxylate transporter 4 (MCT4) and redirecting pyruvate toward oxidation can blunt hypertrophy. We hypothesized that this pathway might be important during I/R. Here, we establish that the pyruvate-lactate axis plays a role in determining myocardial salvage following injury. After I/R, the mitochondrial pyruvate carrier (MPC), required for pyruvate oxidation, is upregulated in the surviving myocardium. In cardiomyocytes lacking the MPC, there was increased cell death and less salvage after I/R, which was associated with an upregulation of MCT4. To determine the importance of pyruvate oxidation, we inhibited MCT4 with a small-molecule drug (VB124) at reperfusion. This strategy normalized reactive oxygen species (ROS), mitochondrial membrane potential (ΔΨ), and Ca2+, increased pyruvate entry to the TCA cycle, increased oxygen consumption, and improved myocardial salvage and functional outcomes following I/R. Our data suggest normalizing pyruvate-lactate metabolism by inhibiting MCT4 is a promising therapy to mitigate I/R injury.
- Published
- 2024
- Full Text
- View/download PDF
293. Lactate transport inhibition therapeutically reprograms fibroblast metabolism in experimental pulmonary fibrosis.
- Author
-
Ziehr DR, Li F, Parnell KM, Krah NM, Leahy KJ, Guillermier C, Varon J, Baron RM, Maron BA, Philp NJ, Hariri LP, Kim EY, Steinhauser ML, Knipe RS, Rutter J, and Oldham WM
- Abstract
Myofibroblast differentiation, essential for driving extracellular matrix synthesis in pulmonary fibrosis, requires increased glycolysis. While glycolytic cells must export lactate, the contributions of lactate transporters to myofibroblast differentiation are unknown. In this study, we investigated how MCT1 and MCT4, key lactate transporters, influence myofibroblast differentiation and experimental pulmonary fibrosis. Our findings reveal that inhibiting MCT1 or MCT4 reduces TGFβ-stimulated pulmonary myofibroblast differentiation in vitro and decreases bleomycin-induced pulmonary fibrosis in vivo . Through comprehensive metabolic analyses, including bioenergetics, stable isotope tracing, metabolomics, and imaging mass spectrometry in both cells and mice, we demonstrate that inhibiting lactate transport enhances oxidative phosphorylation, reduces reactive oxygen species production, and diminishes glucose metabolite incorporation into fibrotic lung regions. Furthermore, we introduce VB253, a novel MCT4 inhibitor, which ameliorates pulmonary fibrosis in both young and aged mice, with comparable efficacy to established antifibrotic therapies. These results underscore the necessity of lactate transport for myofibroblast differentiation, identify MCT1 and MCT4 as promising pharmacologic targets in pulmonary fibrosis, and support further evaluation of lactate transport inhibitors for patients for whom limited therapeutic options currently exist., Competing Interests: W.M.O. has received consulting fees from Nikang Therapeutics outside the scope of this research.J.R. is a consultant and shareholder for Vettore Biosciences.R.S.K. received a Discovery ILD Award from Boehringer Ingelheim and received support through the Partners Drug Development Lab from Bayer Pharmaceuticals, all outside the scope of this research.E.Y.K. received unrelated research funding from Bayer AG, Roche Pharma Research and Early Development, and 10X Genomics. E.Y.K. has a financial interest in Novartis AG unrelated to this work.B.A.M. has received consulting fees from Actelion and Tenax and has performed investigator-initiated research with support from Deerfield, all outside the scope of this research.L.P.H. reports grants from Boehringer Ingelheim Pharmaceuticals, Inc. (BIPI) and has received personal consulting fees from BIPI, Pliant Therapeutics, Clario, and Abbvie Pharmaceuticals.The remaining authors declare that they have no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
294. Enhancing mitochondrial pyruvate metabolism ameliorates myocardial ischemic reperfusion injury.
- Author
-
Visker JR, Cluntun AA, Velasco-Silva JN, Eberhardt DR, Shankar TS, Hamouche R, Ling J, Kwak H, Hillas Y, Aist I, Tseliou E, Navankasattusas S, Chaudhuri D, Ducker GS, Drakos SG, and Rutter J
- Abstract
The established clinical therapy for the treatment of acute myocardial infarction is primary percutaneous coronary intervention (PPCI) to restore blood flow to the ischemic myocardium. PPCI is effective at reperfusing the ischemic myocardium, however the rapid re-introduction of oxygenated blood also can cause ischemia-reperfusion (I/R) injury. Reperfusion injury is the culprit for up to half of the final myocardial damage, but there are no clinical interventions to reduce I/R injury. We previously demonstrated that inhibiting the lactate exporter, monocarboxylate transporter 4 (MCT4), and re-directing pyruvate towards oxidation can blunt isoproterenol-induced hypertrophy. Based on this finding, we hypothesized that the same pathway might be important during I/R. Here, we establish that the pyruvate-lactate metabolic axis plays a critical role in determining myocardial salvage following injury. Post-I/R injury, the mitochondrial pyruvate carrier (MPC), required for pyruvate oxidation, is upregulated in the surviving myocardium following I/R injury. MPC loss in cardiomyocytes caused more cell death with less myocardial salvage, which was associated with an upregulation of MCT4 in the myocardium at risk of injury. We deployed a pharmacological strategy of MCT4 inhibition with a highly selective compound (VB124) at the time of reperfusion. This strategy normalized reactive oxygen species (ROS), mitochondrial membrane potential (Δψ), and Ca
2+ , increased pyruvate entry to TCA cycle, and improved myocardial salvage and functional outcomes following I/R injury. Altogether, our data suggest that normalizing the pyruvate-lactate metabolic axis via MCT4 inhibition is a promising pharmacological strategy to mitigate I/R injury.- Published
- 2024
- Full Text
- View/download PDF
295. Phosphate starvation signaling increases mitochondrial membrane potential through respiration-independent mechanisms.
- Author
-
Ouyang Y, Jeong MY, Cunningham CN, Berg JA, Toshniwal AG, Hughes CE, Seiler K, Van Vranken JG, Cluntun AA, Lam G, Winter JM, Akdogan E, Dove KK, Nowinski SM, West M, Odorizzi G, Gygi SP, Dunn CD, Winge DR, and Rutter J
- Subjects
- Animals, Membrane Potential, Mitochondrial, Adenosine Triphosphate metabolism, Respiration, Mammals metabolism, Phosphates metabolism, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism
- Abstract
Mitochondrial membrane potential directly powers many critical functions of mitochondria, including ATP production, mitochondrial protein import, and metabolite transport. Its loss is a cardinal feature of aging and mitochondrial diseases, and cells closely monitor membrane potential as an indicator of mitochondrial health. Given its central importance, it is logical that cells would modulate mitochondrial membrane potential in response to demand and environmental cues, but there has been little exploration of this question. We report that loss of the Sit4 protein phosphatase in yeast increases mitochondrial membrane potential, both by inducing the electron transport chain and the phosphate starvation response. Indeed, a similarly elevated mitochondrial membrane potential is also elicited simply by phosphate starvation or by abrogation of the Pho85-dependent phosphate sensing pathway. This enhanced membrane potential is primarily driven by an unexpected activity of the ADP/ATP carrier. We also demonstrate that this connection between phosphate limitation and enhancement of mitochondrial membrane potential is observed in primary and immortalized mammalian cells as well as in Drosophila . These data suggest that mitochondrial membrane potential is subject to environmental stimuli and intracellular signaling regulation and raise the possibility for therapeutic enhancement of mitochondrial function even in defective mitochondria., Competing Interests: YO, MJ, CC, JB, AT, CH, KS, JV, AC, GL, JW, EA, KD, SN, MW, GO, SG, CD, DW, JR No competing interests declared, (© 2024, Ouyang et al.)
- Published
- 2024
- Full Text
- View/download PDF
296. Sexual dimorphism of osteoclast reliance on mitochondrial oxidation of energy substrates in the mouse.
- Author
-
Song C, Valeri A, Song F, Ji X, Liao X, Marmo T, Seeley R, Rutter J, and Long F
- Subjects
- Male, Mice, Female, Animals, Sex Characteristics, Mitochondria, Fatty Acids metabolism, Glucose metabolism, Osteoclasts metabolism, Bone Resorption metabolism
- Abstract
Osteoclasts specialize in bone resorption and are critical for bone remodeling. Previous studies have shown that osteoclasts possess abundant mitochondria and derive most energy through oxidative phosphorylation (OXPHOS). However, the energy substrates fueling OXPHOS in osteoclasts remain to be fully defined. Here, we showed that osteoclast differentiation was coupled with increased oxidation of glucose, glutamine, and oleate. Transcriptomic analyses with RNA sequencing revealed marked upregulation of genes participating in OXPHOS and mitochondrial fatty acid oxidation, during osteoclast differentiation. Increased mitochondrial oxidation of long-chain fatty acids was required for osteoclast differentiation in vitro. However, blocking fatty acid oxidation in vivo, by deletion of carnitine palmitoyltransferase 1a (Cpt1a) in osteoclast progenitors, impaired osteoclast formation only in the female mice. The Cpt1a-deficient females were further protected from osteoclast activation by a high-fat diet. The males, on the contrary, exhibited normal bone resorption despite Cpt1a deletion, regardless of the dietary fat content. Moreover, concurrent deletion of mitochondrial pyruvate carrier 1 and Cpt1a, blocking mitochondrial oxidation of both glucose and fatty acids in the osteoclast lineage, failed to impede bone resorption in the males. The study therefore uncovers a female-specific dependence on mitochondrial oxidation of fatty acids and glucose in osteoclasts in vivo.
- Published
- 2023
- Full Text
- View/download PDF
297. Ceramides Increase Fatty Acid Utilization in Intestinal Progenitors to Enhance Stemness and Increase Tumor Risk.
- Author
-
Li Y, Chaurasia B, Rahman MM, Kaddai V, Maschek JA, Berg JA, Wilkerson JL, Mahmassani ZS, Cox J, Wei P, Meikle PJ, Atkinson D, Wang L, Poss AM, Playdon MC, Tippetts TS, Mousa EM, Nittayaboon K, Anandh Babu PV, Drummond MJ, Clevers H, Shayman JA, Hirabayashi Y, Holland WL, Rutter J, Edgar BA, and Summers SA
- Subjects
- Humans, Animals, Mice, Fatty Acids, Sphingolipids metabolism, Serine C-Palmitoyltransferase metabolism, Ceramides metabolism, Adenoma
- Abstract
Background & Aims: Cancers of the alimentary tract, including esophageal adenocarcinomas, colorectal cancers, and cancers of the gastric cardia, are common comorbidities of obesity. Prolonged, excessive delivery of macronutrients to the cells lining the gut can increase one's risk for these cancers by inducing imbalances in the rate of intestinal stem cell proliferation vs differentiation, which can produce polyps and other aberrant growths. We investigated whether ceramides, which are sphingolipids that serve as a signal of nutritional excess, alter stem cell behaviors to influence cancer risk., Methods: We profiled sphingolipids and sphingolipid-synthesizing enzymes in human adenomas and tumors. Thereafter, we manipulated expression of sphingolipid-producing enzymes, including serine palmitoyltransferase (SPT), in intestinal progenitors of mice, cultured organoids, and Drosophila to discern whether sphingolipids altered stem cell proliferation and metabolism., Results: SPT, which diverts dietary fatty acids and amino acids into the biosynthetic pathway that produces ceramides and other sphingolipids, is a critical modulator of intestinal stem cell homeostasis. SPT and other enzymes in the sphingolipid biosynthesis pathway are up-regulated in human intestinal adenomas. They produce ceramides, which serve as prostemness signals that stimulate peroxisome-proliferator activated receptor-α and induce fatty acid binding protein-1. These actions lead to increased lipid utilization and enhanced proliferation of intestinal progenitors., Conclusions: Ceramides serve as critical links between dietary macronutrients, epithelial regeneration, and cancer risk., (Copyright © 2023 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
298. Cardiac gene therapy treats diabetic cardiomyopathy and lowers blood glucose.
- Author
-
Li J, Richmond B, Cluntun AA, Bia R, Walsh MA, Shaw K, Symons JD, Franklin S, Rutter J, Funai K, Shaw RM, and Hong T
- Subjects
- Animals, Mice, Blood Glucose, Calcium, Stroke Volume, Anti-Arrhythmia Agents, Cardiotonic Agents, Myocytes, Cardiac, Adaptor Proteins, Signal Transducing, Amino Acids, Enzyme Inhibitors, Genetic Therapy, Diabetic Cardiomyopathies genetics, Diabetic Cardiomyopathies therapy, Heart Failure therapy, Diabetes Mellitus, Experimental complications, Diabetes Mellitus, Experimental therapy, Diabetes Mellitus, Type 2 complications, Diabetes Mellitus, Type 2 therapy, Hyperglycemia therapy
- Abstract
Diabetic cardiomyopathy, an increasingly global epidemic and a major cause of heart failure with preserved ejection fraction (HFpEF), is associated with hyperglycemia, insulin resistance, and intracardiomyocyte calcium mishandling. Here we identify that, in db/db mice with type 2 diabetes-induced HFpEF, abnormal remodeling of cardiomyocyte transverse-tubule microdomains occurs with downregulation of the membrane scaffolding protein cardiac bridging integrator 1 (cBIN1). Transduction of cBIN1 by AAV9 gene therapy can restore transverse-tubule microdomains to normalize intracellular distribution of calcium-handling proteins and, surprisingly, glucose transporter 4 (GLUT4). Cardiac proteomics revealed that AAV9-cBIN1 normalized components of calcium handling and GLUT4 translocation machineries. Functional studies further identified that AAV9-cBIN1 normalized insulin-dependent glucose uptake in diabetic cardiomyocytes. Phenotypically, AAV9-cBIN1 rescued cardiac lusitropy, improved exercise intolerance, and ameliorated hyperglycemia in diabetic mice. Restoration of transverse-tubule microdomains can improve cardiac function in the setting of diabetic cardiomyopathy and can also improve systemic glycemic control.
- Published
- 2023
- Full Text
- View/download PDF
299. Pyruvate metabolism controls chromatin remodeling during CD4 + T cell activation.
- Author
-
Mocholi E, Russo L, Gopal K, Ramstead AG, Hochrein SM, Vos HR, Geeven G, Adegoke AO, Hoekstra A, van Es RM, Pittol JR, Vastert S, Rutter J, Radstake T, van Loosdregt J, Berkers C, Mokry M, Anderson CC, O'Connell RM, Vaeth M, Ussher J, Burgering BMT, and Coffer PJ
- Subjects
- Humans, Acetyl Coenzyme A metabolism, CD4-Positive T-Lymphocytes metabolism, Histones metabolism, Chromatin Assembly and Disassembly
- Abstract
Upon antigen-specific T cell receptor (TCR) engagement, human CD4
+ T cells proliferate and differentiate, a process associated with rapid transcriptional changes and metabolic reprogramming. Here, we show that the generation of extramitochondrial pyruvate is an important step for acetyl-CoA production and subsequent H3K27ac-mediated remodeling of histone acetylation. Histone modification, transcriptomic, and carbon tracing analyses of pyruvate dehydrogenase (PDH)-deficient T cells show PDH-dependent acetyl-CoA generation as a rate-limiting step during T activation. Furthermore, T cell activation results in the nuclear translocation of PDH and its association with both the p300 acetyltransferase and histone H3K27ac. These data support the tight integration of metabolic and histone-modifying enzymes, allowing metabolic reprogramming to fuel CD4+ T cell activation. Targeting this pathway may provide a therapeutic approach to specifically regulate antigen-driven T cell activation., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
300. The secret life of lactate: A novel cell-cycle regulatory mechanism.
- Author
-
Cluntun AA and Rutter J
- Subjects
- Cell Cycle Checkpoints, Lactic Acid, Mitosis
- Abstract
Recently, Liu et al. uncovered an unexpected L-lactate-Zn
2+ interaction in the active site of the deSUMOylating enzyme SENP1 that triggers a sequence of events that lead to mitotic exit. This study opens the door to new research avenues of metabolite-metal interactions controlling cellular decisions and functions., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)- 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.