ϟ

Bradford G. Hill

Here are all the papers by Bradford G. Hill that you can download and read on OA.mg.
Bradford G. Hill’s last known institution is . Download Bradford G. Hill PDFs here.

Claim this Profile →
DOI: 10.1515/hsz-2012-0198
2012
Cited 396 times
Integration of cellular bioenergetics with mitochondrial quality control and autophagy
Abstract Bioenergetic dysfunction is emerging as a cornerstone for establishing a framework for understanding the pathophysiology of cardiovascular disease, diabetes, cancer and neurodegeneration. Recent advances in cellular bioenergetics have shown that many cells maintain a substantial bioenergetic reserve capacity, which is a prospective index of ‘healthy’ mitochondrial populations. The bioenergetics of the cell are likely regulated by energy requirements and substrate availability. Additionally, the overall quality of the mitochondrial population and the relative abundance of mitochondria in cells and tissues also impinge on overall bioenergetic capacity and resistance to stress. Because mitochondria are susceptible to damage mediated by reactive oxygen/nitrogen and lipid species, maintaining a ‘healthy’ population of mitochondria through quality control mechanisms appears to be essential for cell survival under conditions of pathological stress. Accumulating evidence suggest that mitophagy is particularly important for preventing amplification of initial oxidative insults, which otherwise would further impair the respiratory chain or promote mutations in mitochondrial DNA (mtDNA). The processes underlying the regulation of mitophagy depend on several factors, including the integrity of mtDNA, electron transport chain activity, and the interaction and regulation of the autophagic machinery. The integration and interpretation of cellular bioenergetics in the context of mitochondrial quality control and genetics is the theme of this review.
DOI: 10.1016/j.freeradbiomed.2011.08.005
2011
Cited 377 times
Assessing bioenergetic function in response to oxidative stress by metabolic profiling
It is now clear that mitochondria are an important target for oxidative stress in a broad range of pathologies, including cardiovascular disease, diabetes, neurodegeneration, and cancer. Methods for assessing the impact of reactive species on isolated mitochondria are well established but constrained by the need for large amounts of material to prepare intact mitochondria for polarographic measurements. With the availability of high-resolution polarography and fluorescence techniques for the measurement of oxygen concentration in solution, measurements of mitochondrial function in intact cells can be made. Recently, the development of extracellular flux methods to monitor changes in oxygen concentration and pH in cultures of adherent cells in multiple-sample wells simultaneously has greatly enhanced the ability to measure bioenergetic function in response to oxidative stress. Here we describe these methods in detail using representative cell types from renal, cardiovascular, nervous, and tumorigenic model systems while illustrating the application of three protocols to analyze the bioenergetic response of cells to oxidative stress.
DOI: 10.1016/j.freeradbiomed.2010.01.015
2010
Cited 296 times
Mitochondrial reserve capacity in endothelial cells: The impact of nitric oxide and reactive oxygen species
The endothelium is not considered to be a major energy-requiring organ, but nevertheless endothelial cells have an extensive mitochondrial network. This suggests that mitochondrial function may be important in response to stress and signaling in these cells. In this study, we used extracellular flux analysis to measure mitochondrial function in adherent bovine aortic endothelial cells (BAEC). Under basal conditions, BAEC use only approximately 35% of their maximal respiratory capacity. We calculate that this represents an intermediate respiratory state between States 3 and 4, which we define as State(apparent) equal to 3.64. Interestingly, the apparent respiratory control ratio (maximal mitochondrial oxygen consumption/non-ADP-linked respiration) in these cells is on the order of 23, which is substantially higher than that which is frequently obtained with isolated mitochondria. These results suggest that mitochondria in endothelial cells are highly coupled and possess a considerable bioenergetic reserve. Because endothelial cells are exposed to both reactive oxygen (ROS) and reactive nitrogen species in the course of vascular disease, we hypothesized that this reserve capacity is important in responding to oxidative stress. To test this, we exposed BAEC to NO or ROS alone or in combination. We found that exposure to nontoxic concentrations of NO or low levels of hydrogen peroxide generated from 2,3-dimethoxy-1,4-napthoquinone (DMNQ) had little impact on basal mitochondrial function but both treatments reversibly decreased mitochondrial reserve capacity. However, combined NO and DMNQ treatment resulted in an irreversible loss of reserve capacity and was associated with cell death. These data are consistent with a critical role for the mitochondrial reserve capacity in endothelial cells in responding to oxidative stress.
DOI: 10.1074/jbc.r110.101618
2010
Cited 290 times
What Part of NO Don't You Understand? Some Answers to the Cardinal Questions in Nitric Oxide Biology
Nitric oxide (NO) regulates biological processes through signaling mechanisms that exploit its unique biochemical properties as a free radical. For the last several decades, the key aspects of the chemical properties of NO relevant to biological systems have been defined, but it has been a challenge to assign these to specific cellular processes. Nevertheless, it is now clear that the high affinity of NO for transition metal centers, particularly iron, and the rapid reaction of NO with oxygen-derived free radicals can explain many of its biological and pathological properties. Emerging studies also highlight a growing importance of the secondary metabolites of NO-dependent reactions in the post-translational modification of key metabolic and signaling proteins. In this minireview, we emphasize the current understanding of the biochemistry of NO and place it in a biological context.
DOI: 10.1038/nprot.2014.018
2014
Cited 260 times
Comprehensive measurement of respiratory activity in permeabilized cells using extracellular flux analysis
Extracellular flux (XF) analysis has become a mainstream method for measuring mitochondrial function in cells and tissues. Although this technique is commonly used to measure bioenergetics in intact cells, we outline here a detailed XF protocol for measuring respiration in permeabilized cells. Cells are permeabilized using saponin (SAP), digitonin (DIG) or recombinant perfringolysin O (rPFO) (XF-plasma membrane permeabilizer (PMP) reagent), and they are provided with specific substrates to measure complex I- or complex II-mediated respiratory activity, complex III+IV respiratory activity or complex IV activity. Medium- and long-chain acylcarnitines or glutamine may also be provided for measuring fatty acid (FA) oxidation or glutamine oxidation, respectively. This protocol uses a minimal number of cells compared with other protocols and does not require isolation of mitochondria. The results are highly reproducible, and mitochondria remain well coupled. Collectively, this protocol provides comprehensive and detailed information regarding mitochondrial activity and efficiency, and, after preparative steps, it takes 6-8 h to complete.
DOI: 10.1042/bj20090934
2009
Cited 257 times
Importance of the bioenergetic reserve capacity in response to cardiomyocyte stress induced by 4-hydroxynonenal
Mitochondria play a critical role in mediating the cellular response to oxidants formed during acute and chronic cardiac dysfunction. It is widely assumed that, as cells are subjected to stress, mitochondria are capable of drawing upon a ‘reserve capacity’ which is available to serve the increased energy demands for maintenance of organ function, cellular repair or detoxification of reactive species. This hypothesis further implies that impairment or depletion of this putative reserve capacity ultimately leads to excessive protein damage and cell death. However, it has been difficult to fully evaluate this hypothesis since much of our information about the response of the mitochondrion to oxidative stress derives from studies on mitochondria isolated from their cellular context. Therefore the goal of the present study was to determine whether ‘bioenergetic reserve capacity’ does indeed exist in the intact myocyte and whether it is utilized in response to stress induced by the pathologically relevant reactive lipid species HNE (4-hydroxynonenal). We found that intact rat neonatal ventricular myocytes exhibit a substantial bioenergetic reserve capacity under basal conditions; however, on exposure to pathologically relevant concentrations of HNE, oxygen consumption was increased until this reserve capacity was depleted. Exhaustion of the reserve capacity by HNE treatment resulted in inhibition of respiration concomitant with protein modification and cell death. These data suggest that oxidized lipids could contribute to myocyte injury by decreasing the bioenergetic reserve capacity. Furthermore, these studies demonstrate the utility of measuring the bioenergetic reserve capacity for assessing or predicting the response of cells to stress.
DOI: 10.1161/circresaha.118.312017
2018
Cited 232 times
Metabolic Coordination of Physiological and Pathological Cardiac Remodeling
Metabolic pathways integrate to support tissue homeostasis and to prompt changes in cell phenotype. In particular, the heart consumes relatively large amounts of substrate not only to regenerate ATP for contraction but also to sustain biosynthetic reactions for replacement of cellular building blocks. Metabolic pathways also control intracellular redox state, and metabolic intermediates and end products provide signals that prompt changes in enzymatic activity and gene expression. Mounting evidence suggests that the changes in cardiac metabolism that occur during development, exercise, and pregnancy as well as with pathological stress (eg, myocardial infarction, pressure overload) are causative in cardiac remodeling. Metabolism-mediated changes in gene expression, metabolite signaling, and the channeling of glucose-derived carbon toward anabolic pathways seem critical for physiological growth of the heart, and metabolic inefficiency and loss of coordinated anabolic activity are emerging as proximal causes of pathological remodeling. This review integrates knowledge of different forms of cardiac remodeling to develop general models of how relationships between catabolic and anabolic glucose metabolism may fortify cardiac health or promote (mal)adaptive myocardial remodeling. Adoption of conceptual frameworks based in relational biology may enable further understanding of how metabolism regulates cardiac structure and function.
DOI: 10.1016/j.freeradbiomed.2014.03.025
2014
Cited 203 times
Redox regulation of antioxidants, autophagy, and the response to stress: Implications for electrophile therapeutics
Redox networks in the cell integrate signaling pathways that control metabolism, energetics, cell survival, and death. The physiological second messengers that modulate these pathways include nitric oxide, hydrogen peroxide, and electrophiles. Electrophiles are produced in the cell via both enzymatic and nonenzymatic lipid peroxidation and are also relatively abundant constituents of the diet. These compounds bind covalently to families of cysteine-containing, redox-sensing proteins that constitute the electrophile-responsive proteome, the subproteomes of which are found in localized intracellular domains. These include those proteins controlling responses to oxidative stress in the cytosol—notably the Keap1-Nrf2 pathway, the autophagy-lysosomal pathway, and proteins in other compartments including mitochondria and endoplasmic reticulum. The signaling pathways through which electrophiles function have unique characteristics that could be exploited for novel therapeutic interventions; however, development of such therapeutic strategies has been challenging due to a lack of basic understanding of the mechanisms controlling this form of redox signaling. In this review, we discuss current knowledge of the basic mechanisms of thiol-electrophile signaling and its potential impact on the translation of this important field of redox biology to the clinic. Emerging understanding of thiol-electrophile interactions and redox signaling suggests replacement of the oxidative stress hypothesis with a new redox biology paradigm, which provides an exciting and influential framework for guiding translational research.
DOI: 10.1016/j.freeradbiomed.2014.05.016
2014
Cited 201 times
Regulation of obesity and insulin resistance by nitric oxide
Obesity is a risk factor for developing type 2 diabetes and cardiovascular disease and has quickly become a worldwide pandemic with few tangible and safe treatment options. Although it is generally accepted that the primary cause of obesity is energy imbalance, i.e., the calories consumed are greater than are utilized, understanding how caloric balance is regulated has proven a challenge. Many "distal" causes of obesity, such as the structural environment, occupation, and social influences, are exceedingly difficult to change or manipulate. Hence, molecular processes and pathways more proximal to the origins of obesity-those that directly regulate energy metabolism or caloric intake-seem to be more feasible targets for therapy. In particular, nitric oxide (NO) is emerging as a central regulator of energy metabolism and body composition. NO bioavailability is decreased in animal models of diet-induced obesity and in obese and insulin-resistant patients, and increasing NO output has remarkable effects on obesity and insulin resistance. This review discusses the role of NO in regulating adiposity and insulin sensitivity and places its modes of action into context with the known causes and consequences of metabolic disease.
DOI: 10.1161/circheartfailure.114.001151
2014
Cited 190 times
Metabolomic Analysis of Pressure-Overloaded and Infarcted Mouse Hearts
Background— Cardiac hypertrophy and heart failure are associated with metabolic dysregulation and a state of chronic energy deficiency. Although several disparate changes in individual metabolic pathways have been described, there has been no global assessment of metabolomic changes in hypertrophic and failing hearts in vivo. Hence, we investigated the impact of pressure overload and infarction on myocardial metabolism. Methods and Results— Male C57BL/6J mice were subjected to transverse aortic constriction or permanent coronary occlusion (myocardial infarction [MI]). A combination of LC/MS/MS and GC/MS techniques was used to measure 288 metabolites in these hearts. Both transverse aortic constriction and MI were associated with profound changes in myocardial metabolism affecting up to 40% of all metabolites measured. Prominent changes in branched-chain amino acids were observed after 1 week of transverse aortic constriction and 5 days after MI. Changes in branched-chain amino acids after MI were associated with myocardial insulin resistance. Longer duration of transverse aortic constriction and MI led to a decrease in purines, acylcarnitines, fatty acids, and several lysolipid and sphingolipid species but a marked increase in pyrimidines as well as ascorbate, heme, and other indices of oxidative stress. Cardiac remodeling and contractile dysfunction in hypertrophied hearts were associated with large increases in myocardial, but not plasma, levels of the polyamines putrescine and spermidine as well as the collagen breakdown product prolylhydroxyproline. Conclusions— These findings reveal extensive metabolic remodeling common to both hypertrophic and failing hearts that are indicative of extracellular matrix remodeling, insulin resistance and perturbations in amino acid, and lipid and nucleotide metabolism.
DOI: 10.1042/bj20121344
2013
Cited 181 times
PDGF-mediated autophagy regulates vascular smooth muscle cell phenotype and resistance to oxidative stress
Vascular injury and chronic arterial diseases result in exposure of VSMCs (vascular smooth muscle cells) to increased concentrations of growth factors. The mechanisms by which growth factors trigger VSMC phenotype transitions remain unclear. Because cellular reprogramming initiated by growth factors requires not only the induction of genes involved in cell proliferation, but also the removal of contractile proteins, we hypothesized that autophagy is an essential modulator of VSMC phenotype. Treatment of VSMCs with PDGF (platelet-derived growth factor)-BB resulted in decreased expression of the contractile phenotype markers calponin and α-smooth muscle actin and up-regulation of the synthetic phenotype markers osteopontin and vimentin. Autophagy, as assessed by LC3 (microtubule-associated protein light chain 3 α; also known as MAP1LC3A)-II abundance, LC3 puncta formation and electron microscopy, was activated by PDGF exposure. Inhibition of autophagy with 3-methyladenine, spautin-1 or bafilomycin stabilized the contractile phenotype. In particular, spautin-1 stabilized α-smooth muscle cell actin and calponin in PDGF-treated cells and prevented actin filament disorganization, diminished production of extracellular matrix, and abrogated VSMC hyperproliferation and migration. Treatment of cells with PDGF prevented protein damage and cell death caused by exposure to the lipid peroxidation product 4-hydroxynonenal. The results of the present study demonstrate a distinct form of autophagy induced by PDGF that is essential for attaining the synthetic phenotype and for survival under the conditions of high oxidative stress found to occur in vascular lesions.
DOI: 10.1038/onc.2012.635
2013
Cited 173 times
Control of glutamine metabolism by the tumor suppressor Rb
Retinoblastoma (Rb) protein is a tumor suppressor that is dysregulated in a majority of human cancers. Rb functions to inhibit cell cycle progression in part by directly disabling the E2F family of cell cycle-promoting transcription factors. Because the de novo synthesis of multiple glutamine-derived anabolic precursors is required for cell cycle progression, we hypothesized that Rb also may directly regulate proteins involved in glutamine metabolism. We examined glutamine metabolism in mouse embryonic fibroblasts (MEFs) isolated from mice that have triple knock-outs (TKO) of all three Rb family members (Rb-1, Rbl1 and Rbl2) and found that loss of global Rb function caused a marked increase in (13)C-glutamine uptake and incorporation into glutamate and tricarboxylic acid cycle (TCA) intermediates in part via upregulated expression of the glutamine transporter ASCT2 and the activity of glutaminase 1 (GLS1). The Rb-controlled transcription factor E2F-3 altered glutamine uptake by direct regulation of ASCT2 mRNA and protein expression, and E2F-3 was observed to associate with the ASCT2 promoter. We next examined the functional consequences of the observed increase in glutamine uptake and utilization and found that glutamine exposure potently increased oxygen consumption, whereas glutamine deprivation selectively decreased ATP concentration in the Rb TKO MEFs but not the wild-type (WT) MEFs. In addition, TKO MEFs exhibited elevated production of glutathione from exogenous glutamine and had increased expression of gamma-glutamylcysteine ligase relative to WT MEFs. Importantly, this metabolic shift towards glutamine utilization was required for the proliferation of Rb TKO MEFs but not for the proliferation of the WT MEFs. Last, addition of the TCA cycle intermediate α-ketoglutarate to the Rb TKO MEFs reversed the inhibitory effects of glutamine deprivation on ATP, GSH levels and viability. Taken together, these studies demonstrate that the Rb/E2F cascade directly regulates a major energetic and anabolic pathway that is required for neoplastic growth.
DOI: 10.1172/jci131335
2020
Cited 114 times
Transcription factor c-Maf is a checkpoint that programs macrophages in lung cancer
Macrophages have been linked to tumor initiation, progression, metastasis, and treatment resistance.However, the transcriptional regulation of macrophages driving the protumor function remains elusive.Here, we demonstrate that the transcription factor c-Maf is a critical controller for immunosuppressive macrophage polarization and function in cancer.c-Maf controls many M2-related genes and has direct binding sites within a conserved noncoding sequence of the Csf-1r gene and promotes M2-like macrophage-mediated T cell suppression and tumor progression.c-Maf also serves as a metabolic checkpoint regulating the TCA cycle and UDP-GlcNAc biosynthesis, thus promoting M2-like macrophage polarization and activation.Additionally, c-Maf is highly expressed in tumor-associated macrophages (TAMs) and regulates TAM immunosuppressive function.Deletion of c-Maf specifically in myeloid cells results in reduced tumor burden with enhanced antitumor T cell immunity.Inhibition of c-Maf partly overcomes resistance to anti-PD-1 therapy in a subcutaneous LLC tumor model.Similarly, c-Maf is expressed in human M2 and tumor-infiltrating macrophages/monocytes as well as circulating monocytes of human non-small cell lung carcinoma (NSCLC) patients and critically regulates their immunosuppressive activity.The natural compound β-glucan downregulates c-Maf expression on macrophages, leading to enhanced antitumor immunity in mice.These findings establish a paradigm for immunosuppressive macrophage polarization and transcriptional regulation by c-Maf and suggest that c-Maf is a potential target for effective tumor immunotherapy.
DOI: 10.1161/circulationaha.107.730515
2008
Cited 227 times
Cardioprotection by <i>N</i> -Acetylglucosamine Linkage to Cellular Proteins
Background— The modification of proteins with O -linked β- N -acetylglucosamine ( O -GlcNAc) represents a key posttranslational modification that modulates cellular function. Previous data suggest that O -GlcNAc may act as an intracellular metabolic or stress sensor, linking glucose metabolism to cellular function. Considering this, we hypothesized that augmentation of O -GlcNAc levels represents an endogenously recruitable mechanism of cardioprotection. Methods and Results— In mouse hearts subjected to in vivo ischemic preconditioning, O -GlcNAc levels were significantly elevated. Pharmacological augmentation of O -GlcNAc levels in vivo was sufficient to reduce myocardial infarct size. We investigated the influence of O -GlcNAc levels on cardiac injury at the cellular level. Lethal oxidant stress of cardiac myocytes produced a time-dependent loss of cellular O -GlcNAc levels. This pathological response was largely reversible by pharmacological augmentation of O -GlcNAc levels and was associated with improved cardiac myocyte survival. The diminution of O -GlcNAc levels occurred synchronously with the loss of mitochondrial membrane potential in isolated cardiac myocytes. Pharmacological enhancement of O -GlcNAc levels attenuated the loss of mitochondrial membrane potential. Proteomic analysis identified voltage-dependent anion channel as a potential target of O -GlcNAc modification. Mitochondria isolated from adult mouse hearts with elevated O -GlcNAc levels had more O -GlcNAc–modified voltage-dependent anion channel and were more resistant to calcium-induced swelling than cardiac mitochondria from vehicle mice. Conclusions— O -GlcNAc signaling represents a unique endogenously recruitable mechanism of cardioprotection that may involve direct modification of mitochondrial proteins critical for survival such as voltage-dependent anion channel.
DOI: 10.1016/j.redox.2012.10.003
2013
Cited 162 times
Oxidized lipids activate autophagy in a JNK-dependent manner by stimulating the endoplasmic reticulum stress response
Excessive production of unsaturated aldehydes from oxidized lipoproteins and membrane lipids is a characteristic feature of cardiovascular disease. Our previous studies show that unsaturated lipid peroxidation-derived aldehydes such as 4-hydroxy-trans-2-nonenal (HNE) promote autophagy in rat aortic smooth muscle cells (RASMC). In this study, we examined the mechanism by which HNE induces autophagy. Exposure of RASMC to HNE led to the modification of several proteins, most of which were identified by mass spectrometry and confocal microscopy to be localized to the endoplasmic reticulum (ER). HNE stimulated the phosphorylation of PKR-like ER kinase and eukaryotic initiation factor 2α and increased heme oxygenase-1 (HO-1) abundance. HNE treatment also increased LC3-II formation and the phosphorylation of JNK and p38. Pharmacological inhibition of JNK, but not p38, prevented HNE-induced HO-1 expression and LC3-II formation. Inhibition of JNK increased cell death in HNE-treated cells. Pretreatment with the chemical chaperone phenylbutryic acid prevented LC3-II formation as well as JNK phosphorylation and HO-1 induction. Taken together, these data suggest that autophagic responses triggered by unsaturated aldehydes could be attributed, in part, to ER stress, which stimulates autophagy by a JNK-dependent mechanism and promotes cell survival during oxidative stress.
DOI: 10.1042/bj20071063
2008
Cited 160 times
Unsaturated lipid peroxidation-derived aldehydes activate autophagy in vascular smooth-muscle cells
Proteins modified by aldehydes generated from oxidized lipids accumulate in cells during oxidative stress and are commonly detected in diseased or aged tissue. The mechanisms by which cells remove aldehyde-adducted proteins, however, remain unclear. Here, we report that products of lipid peroxidation such as 4-HNE (4-hydroxynonenal) and acrolein activate autophagy in rat aortic smooth-muscle cells in culture. Exposure to 4-HNE led to the modification of several proteins, as detected by anti-protein–4-HNE antibodies or protein-bound radioactivity in [3H]4-HNE-treated cells. The 4-HNE-modified proteins were gradually removed from cells. The removal of 4-HNE-modified proteins was not affected by the oxidized protein hydrolase inhibitor, acetyl leucine chloromethyl ketone, or lactacystin, although it was significantly decreased by PSI (proteasome inhibitor I), the lysosome/proteasome inhibitor MG-132 (carbobenzoxy-L-leucyl-L-leucyl-leucinal), insulin or the autophagy inhibitor 3-MA (3-methyladenine). Pre-incubation of cells with rapamycin accelerated the removal of 4-HNE-modified proteins. Treatment with 4-HNE, nonenal and acrolein, but not nonanal or POVPC (1-palmitoyl-2-oxovaleroyl phosphatidyl choline), caused a robust increase in LC3-II (microtubule-associated protein 1 light chain 3-II) formation, which was increased also by rapamycin, but prevented by insulin. Electron micrographs of 4-HNE-treated cells showed extensive vacuolization, pinocytic body formation, crescent-shaped phagophores, and multilamellar vesicles. Treatment with 3-MA and MG-132, but not proteasome-specific inhibitors, induced cell death in 4-HNE-treated cells. Collectively, these results show that lipid peroxidation-derived aldehydes stimulate autophagy, which removes aldehyde-modified proteins, and that inhibition of autophagy precipitates cell death in aldehyde-treated cells. Autophagy may be an important mechanism for the survival of arterial smooth-muscle cells under conditions associated with excessive lipid peroxidation.
DOI: 10.1152/ajpheart.01056.2008
2009
Cited 137 times
Protein<i>O</i>-GlcNAcylation: a new signaling paradigm for the cardiovascular system
The posttranslational modification of serine and threonine residues of nuclear and cytoplasmic proteins by the O-linked attachment of the monosaccharide β- N-acetylglucosamine ( O-GlcNAc) is a highly dynamic and ubiquitous protein modification. Protein O-GlcNAcylation is rapidly emerging as a key regulator of critical biological processes including nuclear transport, translation and transcription, signal transduction, cytoskeletal reorganization, proteasomal degradation, and apoptosis. Increased levels of O-GlcNAc have been implicated as a pathogenic contributor to glucose toxicity and insulin resistance, which are both major hallmarks of diabetes mellitus and diabetes-related cardiovascular complications. Conversely, there is a growing body of data demonstrating that the acute activation of O-GlcNAc levels is an endogenous stress response designed to enhance cell survival. Reports on the effect of altered O-GlcNAc levels on the heart and cardiovascular system have been growing rapidly over the past few years and have implicated a role for O-GlcNAc in contributing to the adverse effects of diabetes on cardiovascular function as well as mediating the response to ischemic injury. Here, we summarize our present understanding of protein O-GlcNAcylation and its effect on the regulation of cardiovascular function. We examine the pathways regulating protein O-GlcNAcylation and discuss, in more detail, our understanding of the role of O-GlcNAc in both mediating the adverse effects of diabetes as well as its role in mediating cellular protective mechanisms in the cardiovascular system. In addition, we also explore the parallels between O-GlcNAc signaling and redox signaling, as an alternative paradigm for understanding the role of O-GlcNAcylation in regulating cell function.
DOI: 10.1152/ajpendo.00271.2013
2014
Cited 133 times
Metabolic remodeling of white adipose tissue in obesity
Adipose tissue metabolism is a critical regulator of adiposity and whole body energy expenditure; however, metabolic changes that occur in white adipose tissue (WAT) with obesity remain unclear. The purpose of this study was to understand the metabolic and bioenergetic changes occurring in WAT with obesity. Wild-type (C57BL/6J) mice fed a high-fat diet (HFD) showed significant increases in whole body adiposity, had significantly lower V̇o 2 , V̇co 2 , and respiratory exchange ratios, and demonstrated worsened glucose and insulin tolerance compared with low-fat-fed mice. Metabolomic analysis of WAT showed marked changes in lipid, amino acid, carbohydrate, nucleotide, and energy metabolism. Tissue levels of succinate and malate were elevated, and metabolites that could enter the Krebs cycle via anaplerosis were mostly diminished in high-fat-fed mice, suggesting altered mitochondrial metabolism. Despite no change in basal oxygen consumption or mitochondrial DNA abundance, citrate synthase activity was decreased by more than 50%, and responses to FCCP were increased in WAT from mice fed a high-fat diet. Moreover, Pgc1a was downregulated and Cox7a1 upregulated after 6 wk of HFD. After 12 wk of high-fat diet, the abundance of several proteins in the mitochondrial respiratory chain or matrix was diminished. These changes were accompanied by increased Parkin and Pink1, decreased p62 and LC3-I, and ultrastructural changes suggestive of autophagy and mitochondrial remodeling. These studies demonstrate coordinated restructuring of metabolism and autophagy that could contribute to the hypertrophy and whitening of adipose tissue in obesity.
DOI: 10.1016/j.cbi.2010.12.002
2011
Cited 132 times
Bioenergetic function in cardiovascular cells: The importance of the reserve capacity and its biological regulation
The ability of the cell to generate sufficient energy through oxidative phosphorylation and to maintain healthy pools of mitochondria are critical for survival and maintenance of normal biological function, especially during periods of increased oxidative stress. Mitochondria in most cardiovascular cells function at a basal level that only draws upon a small fraction of the total bioenergetic capability of the organelle; the apparent respiratory state of mitochondria in these cells is often close to state 4. The difference between the basal and maximal activity, equivalent to state 3, of the respiratory chain is called the reserve capacity. We hypothesize that the reserve capacity serves the increased energy demands for maintenance of organ function and cellular repair. However, the factors that determine the volume of the reserve capacity and its relevance to biology are not well understood. In this study, we first examined whether responses to 4-hydroxynonenal (HNE), a lipid peroxidation product found in atherosclerotic lesions and the diseased heart, differ between vascular smooth muscle cells, adult mouse cardiomyocytes, and rat neonatal cardiomyocytes. In both types of cardiomyocytes, oxygen consumption increased after HNE treatment, while oxygen consumption in smooth muscle cells decreased. The increase in oxygen consumption in cardiomyocytes decreased the reserve capacity and shifted the apparent respiratory state closer to state 3. Neonatal rat cardiomyocytes respiring on pyruvate alone had a fourfold higher reserve capacity than cells with glucose as the sole substrate, and these cells were more resistant to mitochondrial dysfunction induced by 4-HNE. The integration of the concepts of reserve capacity and state-apparent are discussed along with the proposal of two potential models by which mitochondria respond to stress.
DOI: 10.1161/circresaha.112.266395
2012
Cited 131 times
Overexpression of Endothelial Nitric Oxide Synthase Prevents Diet-Induced Obesity and Regulates Adipocyte Phenotype
Endothelial dysfunction is a characteristic feature of diabetes and obesity in animal models and humans. Deficits in nitric oxide production by endothelial nitric oxide synthase (eNOS) are associated with insulin resistance, which is exacerbated by high-fat diet. Nevertheless, the metabolic effects of increasing eNOS levels have not been studied.The current study was designed to test whether overexpression of eNOS would prevent diet-induced obesity and insulin resistance.In db/db mice and in high-fat diet-fed wild-type C57BL/6J mice, the abundance of eNOS protein in adipose tissue was decreased without significant changes in eNOS levels in skeletal muscle or aorta. Mice overexpressing eNOS (eNOS transgenic mice) were resistant to diet-induced obesity and hyperinsulinemia, although systemic glucose intolerance remained largely unaffected. In comparison with wild-type mice, high-fat diet-fed eNOS transgenic mice displayed a higher metabolic rate and attenuated hypertrophy of white adipocytes. Overexpression of eNOS did not affect food consumption or diet-induced changes in plasma cholesterol or leptin levels, yet plasma triglycerides and fatty acids were decreased. Metabolomic analysis of adipose tissue indicated that eNOS overexpression primarily affected amino acid and lipid metabolism; subpathway analysis suggested changes in fatty acid oxidation. In agreement with these findings, adipose tissue from eNOS transgenic mice showed higher levels of PPAR-α and PPAR-γ gene expression, elevated abundance of mitochondrial proteins, and a higher rate of oxygen consumption.These findings demonstrate that increased eNOS activity prevents the obesogenic effects of high-fat diet without affecting systemic insulin resistance, in part, by stimulating metabolic activity in adipose tissue.
DOI: 10.1016/j.redox.2013.10.011
2013
Cited 129 times
Mitochondrial fission induced by platelet-derived growth factor regulates vascular smooth muscle cell bioenergetics and cell proliferation
Vascular smooth muscle cells (VSMCs) develop a highly proliferative and synthetic phenotype in arterial diseases. Because such phenotypic changes are likely integrated with the energetic state of the cell, we hypothesized that changes in cellular metabolism regulate VSMC plasticity. VSMCs were exposed to platelet-derived growth factor-BB (PDGF) and changes in mitochondrial morphology, proliferation, contractile protein expression, and mitochondrial metabolism were examined. Exposure of VSMCs to PDGF resulted in mitochondrial fragmentation and a 50% decrease in the abundance of mitofusin 2. Synthetic VSMCs demonstrated a 20% decrease in glucose oxidation, which was accompanied by an increase in fatty acid oxidation. Results of mitochondrial function assays in permeabilized cells showed few changes due to PDGF treatment in mitochondrial respiratory chain capacity and coupling. Treatment of VSMCs with Mdivi-1-an inhibitor of mitochondrial fission-inhibited PDGF-induced mitochondrial fragmentation by 50% and abolished increases in cell proliferation; however, it failed to prevent PDGF-mediated activation of autophagy and removal of contractile proteins. In addition, treatment with Mdivi-1 reversed changes in fatty acid and glucose oxidation associated with the synthetic phenotype. These results suggest that changes in mitochondrial morphology and bioenergetics underlie the hyperproliferative features of the synthetic VSMC phenotype, but do not affect the degradation of contractile proteins. Mitochondrial fragmentation occurring during the transition to the synthetic phenotype could be a therapeutic target for hyperproliferative vascular disorders.
DOI: 10.1161/circulationaha.117.028274
2017
Cited 111 times
Exercise-Induced Changes in Glucose Metabolism Promote Physiological Cardiac Growth
Exercise promotes metabolic remodeling in the heart, which is associated with physiological cardiac growth; however, it is not known whether or how physical activity-induced changes in cardiac metabolism cause myocardial remodeling. In this study, we tested whether exercise-mediated changes in cardiomyocyte glucose metabolism are important for physiological cardiac growth.We used radiometric, immunologic, metabolomic, and biochemical assays to measure changes in myocardial glucose metabolism in mice subjected to acute and chronic treadmill exercise. To assess the relevance of changes in glycolytic activity, we determined how cardiac-specific expression of mutant forms of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase affect cardiac structure, function, metabolism, and gene programs relevant to cardiac remodeling. Metabolomic and transcriptomic screenings were used to identify metabolic pathways and gene sets regulated by glycolytic activity in the heart.Exercise acutely decreased glucose utilization via glycolysis by modulating circulating substrates and reducing phosphofructokinase activity; however, in the recovered state following exercise adaptation, there was an increase in myocardial phosphofructokinase activity and glycolysis. In mice, cardiac-specific expression of a kinase-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase transgene (GlycoLo mice) lowered glycolytic rate and regulated the expression of genes known to promote cardiac growth. Hearts of GlycoLo mice had larger myocytes, enhanced cardiac function, and higher capillary-to-myocyte ratios. Expression of phosphatase-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase in the heart (GlycoHi mice) increased glucose utilization and promoted a more pathological form of hypertrophy devoid of transcriptional activation of the physiological cardiac growth program. Modulation of phosphofructokinase activity was sufficient to regulate the glucose-fatty acid cycle in the heart; however, metabolic inflexibility caused by invariantly low or high phosphofructokinase activity caused modest mitochondrial damage. Transcriptomic analyses showed that glycolysis regulates the expression of key genes involved in cardiac metabolism and remodeling.Exercise-induced decreases in glycolytic activity stimulate physiological cardiac remodeling, and metabolic flexibility is important for maintaining mitochondrial health in the heart.
DOI: 10.1016/j.yjmcc.2011.07.009
2012
Cited 110 times
Protein S-glutathiolation: Redox-sensitive regulation of protein function
Reversible protein S-glutathiolation has emerged as an important mechanism of post-translational modification. Under basal conditions several proteins remain adducted to glutathione, and physiological glutathiolation of proteins has been shown to regulate protein function. Enzymes that promote glutathiolation (e.g., glutathione-S-transferase-P) or those that remove glutathione from proteins (e.g., glutaredoxin) have been identified. Modification by glutathione has been shown to affect protein catalysis, ligand binding, oligomerization and protein-protein interactions. Conditions associated with oxidative or nitrosative stress, such as ischemia-reperfusion, hypertension and tachycardia increase protein glutathiolation via changes in the glutathione redox status (GSH/GSSG) or through the formation of sulfenic acid (SOH) or nitrosated (SNO) cysteine intermediates. These "activated" thiols promote reversible S-glutathiolation of key proteins involved in cell signaling, energy production, ion transport, and cell death. Hence, S-glutathiolation is ideally suited for integrating and mounting fine-tuned responses to changes in the redox state. S-glutathiolation also provides a temporary glutathione "cap" to protect protein thiols from irreversible oxidation and it could be an important mechanism of protein "encryption" to maintain proteins in a functionally silent state until they are needed during conditions of stress. Current evidence suggests that the glutathiolation-deglutathiolation cycle integrates and interacts with other post-translational mechanisms to regulate signal transduction, metabolism, inflammation, and apoptosis. This article is part of a Special Section entitled "Post-translational Modification."
DOI: 10.1074/jbc.m111.320416
2012
Cited 105 times
Lipid Peroxidation Product 4-Hydroxy-trans-2-nonenal Causes Endothelial Activation by Inducing Endoplasmic Reticulum Stress
Lipid peroxidation products, such as 4-hydroxy-<i>trans</i>-2-nonenal (HNE), cause endothelial activation, and they increase the adhesion of the endothelium to circulating leukocytes. Nevertheless, the mechanisms underlying these effects remain unclear. We observed that in HNE-treated human umbilical vein endothelial cells, some of the protein-HNE adducts colocalize with the endoplasmic reticulum (ER) and that HNE forms covalent adducts with several ER chaperones that assist in protein folding. We also found that at concentrations that did not induce apoptosis or necrosis, HNE activated the unfolded protein response, leading to an increase in XBP-1 splicing, phosphorylation of protein kinase-like ER kinase and eukaryotic translation initiation factor 2α, and the induction of ATF3 and ATF4. This increase in eukaryotic translation initiation factor 2α phosphorylation was prevented by transfection with protein kinase-like ER kinase siRNA. Treatment with HNE increased the expression of the ER chaperones, GRP78 and HERP. Exposure to HNE led to a depletion of reduced glutathione and an increase in the production of reactive oxygen species (ROS); however, glutathione depletion and ROS production by <i>tert</i>-butyl-hydroperoxide did not trigger the unfolded protein response. Pretreatment with a chemical chaperone, phenylbutyric acid, or adenoviral transfection with ATF6 attenuated HNE-induced monocyte adhesion and IL-8 induction. Moreover, phenylbutyric acid and taurine-conjugated ursodeoxycholic acid attenuated HNE-induced leukocyte rolling and their firm adhesion to the endothelium in rat cremaster muscle. These data suggest that endothelial activation by HNE is mediated in part by ER stress, induced by mechanisms independent of ROS production or glutathione depletion. The induction of ER stress may be a significant cause of vascular inflammation induced by products of oxidized lipids.
DOI: 10.1038/s41467-019-12103-x
2019
Cited 100 times
Mitochondrial calcium exchange links metabolism with the epigenome to control cellular differentiation
Fibroblast to myofibroblast differentiation is crucial for the initial healing response but excessive myofibroblast activation leads to pathological fibrosis. Therefore, it is imperative to understand the mechanisms underlying myofibroblast formation. Here we report that mitochondrial calcium (mCa2+) signaling is a regulatory mechanism in myofibroblast differentiation and fibrosis. We demonstrate that fibrotic signaling alters gating of the mitochondrial calcium uniporter (mtCU) in a MICU1-dependent fashion to reduce mCa2+ uptake and induce coordinated changes in metabolism, i.e., increased glycolysis feeding anabolic pathways and glutaminolysis yielding increased α-ketoglutarate (αKG) bioavailability. mCa2+-dependent metabolic reprogramming leads to the activation of αKG-dependent histone demethylases, enhancing chromatin accessibility in loci specific to the myofibroblast gene program, resulting in differentiation. Our results uncover an important role for the mtCU beyond metabolic regulation and cell death and demonstrate that mCa2+ signaling regulates the epigenome to influence cellular differentiation.
DOI: 10.1016/j.freeradbiomed.2013.08.003
2013
Cited 89 times
Implications of autophagy for vascular smooth muscle cell function and plasticity
Vascular smooth muscle cells (VSMCs) are fundamental in regulating blood pressure and distributing oxygen and nutrients to peripheral tissues. They also possess remarkable plasticity, with the capacity to switch to synthetic, macrophage-like, or osteochondrogenic phenotypes when cued by external stimuli. In arterial diseases such as atherosclerosis and restenosis, this plasticity seems to be critical and, depending on the disease context, can be deleterious or beneficial. Therefore, understanding the mechanisms regulating VSMC phenotype and survival is essential for developing new therapies for vascular disease as well as understanding how secondary complications due to surgical interventions develop. In this regard, the cellular process of autophagy is increasingly being recognized as a major player in vascular biology and a critical determinant of VSMC phenotype and survival. Although autophagy was identified in lesional VSMCs in the 1960s, our understanding of the implications of autophagy in arterial diseases and the stimuli promoting its activation in VSMCs is only now being elucidated. In this review, we highlight the evidence for autophagy occurring in VSMCs in vivo, elaborate on the stimuli and processes regulating autophagy, and discuss the current understanding of the role of autophagy in vascular disease.
DOI: 10.1161/circresaha.119.314996
2019
Cited 62 times
Physiological Biomimetic Culture System for Pig and Human Heart Slices
Rationale: Preclinical testing of cardiotoxicity and efficacy of novel heart failure therapies faces a major limitation: the lack of an in situ culture system that emulates the complexity of human heart tissue and maintains viability and functionality for a prolonged time. Objective: To develop a reliable, easily reproducible, medium-throughput method to culture pig and human heart slices under physiological conditions for a prolonged period of time. Methods and Results: Here, we describe a novel, medium-throughput biomimetic culture system that maintains viability and functionality of human and pig heart slices (300 µm thickness) for 6 days in culture. We optimized the medium and culture conditions with continuous electrical stimulation at 1.2 Hz and oxygenation of the medium. Functional viability of these slices over 6 days was confirmed by assessing their calcium homeostasis, twitch force generation, and response to β-adrenergic stimulation. Temporal transcriptome analysis using RNAseq at day 2, 6, and 10 in culture confirmed overall maintenance of normal gene expression for up to 6 days, while over 500 transcripts were differentially regulated after 10 days. Electron microscopy demonstrated intact mitochondria and Z-disc ultra-structures after 6 days in culture under our optimized conditions. This biomimetic culture system was successful in keeping human heart slices completely viable and functionally and structurally intact for 6 days in culture. We also used this system to demonstrate the effects of a novel gene therapy approach in human heart slices. Furthermore, this culture system enabled the assessment of contraction and relaxation kinetics on isolated single myofibrils from heart slices after culture. Conclusions: We have developed and optimized a reliable medium-throughput culture system for pig and human heart slices as a platform for testing the efficacy of novel heart failure therapeutics and reliable testing of cardiotoxicity in a 3-dimensional heart model.
DOI: 10.1161/circulationaha.121.057641
2022
Cited 37 times
Transient Cell Cycle Induction in Cardiomyocytes to Treat Subacute Ischemic Heart Failure
The regenerative capacity of the heart after myocardial infarction is limited. Our previous study showed that ectopic introduction of 4 cell cycle factors (4F; CDK1 [cyclin-dependent kinase 1], CDK4 [cyclin-dependent kinase 4], CCNB [cyclin B1], and CCND [cyclin D1]) promotes cardiomyocyte proliferation in 15% to 20% of infected cardiomyocytes in vitro and in vivo and improves cardiac function after myocardial infarction in mice.Using temporal single-cell RNA sequencing, we aimed to identify the necessary reprogramming stages during the forced cardiomyocyte proliferation with 4F on a single cell basis. Using rat and pig models of ischemic heart failure, we aimed to start the first preclinical testing to introduce 4F gene therapy as a candidate for the treatment of ischemia-induced heart failure.Temporal bulk and single-cell RNA sequencing and further biochemical validations of mature human induced pluripotent stem cell-derived cardiomyocytes treated with either LacZ or 4F adenoviruses revealed full cell cycle reprogramming in 15% of the cardiomyocyte population at 48 hours after infection with 4F, which was associated mainly with sarcomere disassembly and metabolic reprogramming (n=3/time point/group). Transient overexpression of 4F, specifically in cardiomyocytes, was achieved using a polycistronic nonintegrating lentivirus (NIL) encoding 4F; each is driven by a TNNT2 (cardiac troponin T isoform 2) promoter (TNNT2-4Fpolycistronic-NIL). TNNT2-4Fpolycistronic-NIL or control virus was injected intramyocardially 1 week after myocardial infarction in rats (n=10/group) or pigs (n=6-7/group). Four weeks after injection, TNNT2-4Fpolycistronic-NIL-treated animals showed significant improvement in left ventricular ejection fraction and scar size compared with the control virus-treated animals. At 4 months after treatment, rats that received TNNT2-4Fpolycistronic-NIL still showed a sustained improvement in cardiac function and no obvious development of cardiac arrhythmias or systemic tumorigenesis (n=10/group).This study provides mechanistic insights into the process of forced cardiomyocyte proliferation and advances the clinical feasibility of this approach by minimizing the oncogenic potential of the cell cycle factors owing to the use of a novel transient and cardiomyocyte-specific viral construct.
DOI: 10.1161/circulationaha.108.791533
2008
Cited 113 times
Cardiac Myocyte–Specific Expression of Inducible Nitric Oxide Synthase Protects Against Ischemia/Reperfusion Injury by Preventing Mitochondrial Permeability Transition
Inducible nitric oxide synthase (iNOS) is an obligatory mediator of the late phase of ischemic preconditioning, but the mechanisms of its cardioprotective actions are unknown. In addition, it remains unclear whether sustained elevation of iNOS in myocytes provides chronic protection against ischemia/reperfusion injury.Constitutive overexpression of iNOS in transgenic mice (alpha-myosin heavy chain promoter) did not induce contractile dysfunction and did not affect mitochondrial respiration or biogenesis, but it profoundly decreased infarct size in mice subjected to 30 minutes of coronary occlusion and 24 hours of reperfusion. In comparison with wild-type hearts, isolated iNOS-transgenic hearts subjected to ischemia for 30 minutes followed by 40 minutes of reperfusion displayed better contractile recovery, smaller infarct size, and less mitochondrial entrapment of 2-deoxy-[(3)H]-glucose. Reperfusion-induced loss of NAD(+) and mitochondrial release of cytochrome c were attenuated in iNOS-transgenic hearts, indicating reduced mitochondrial permeability transition. The NO donor NOC-22 prevented permeability transition in isolated mitochondria, and mitochondrial permeability transition-induced NAD(+) loss was decreased in wild-type but not iNOS-null mice treated with the NO donor diethylene triamine/NO 24 hours before ischemia and reperfusion ex vivo. iNOS-mediated cardioprotection was not abolished by atractyloside. Reperfusion-induced production of oxygen-derived free radicals (measured by electron paramagnetic resonance spectroscopy) was attenuated in iNOS-transgenic hearts and was increased in wild-type hearts treated with the mitochondrial permeability transition inhibitor cyclosporin A.Cardiomyocyte-restricted expression of iNOS provides sustained cardioprotection. This cardioprotection is associated with a decrease in reperfusion-induced oxygen radicals and inhibition of mitochondrial swelling and permeability transition.
DOI: 10.1096/fj.06-5843fje
2006
Cited 113 times
Protein glutathiolation by nitric oxide: an intracellular mechanism regulating redox protein modification
This study was designed to examine whether NO regulates protein glutathiolation. Exposure to NO donors increased protein glutathiolation in COS-7 or rat aortic smooth muscle cells as detected by anti-protein glutathione (GSH) antibodies. This process was reversible and saturable. Stimulation with acetylcholine (ACh) increased protein glutathiolation in isolated rat aortic rings. This was prevented by inhibiting endothelial NO synthase (eNOS). In ACh-treated rings, proteins showing positive immunoreactivity with the anti-PSSG antibody (Ab) were identified by matrix assisted laser desorption-time-of-flight mass spectrometry to be actin, vimentin, and heat shock protein 70. Purified actin was more readily glutathiolated by S-nitrosoglutathione than by oxidized GSH as determined by electrospray-ionization mass spectrometry, and nitrosylated actin was glutathiolated by reduced GSH. Relative to wild-type (WT) mice, increased protein glutathiolation was observed in hearts of mice with cardiac-specific expression of inducible NO synthase (iNOS). Proteins immunoprecipitated from transgenic hearts revealed GSH-adducted peptides corresponding to adenine nucleotide translocator and the alpha-subunit of F1F0ATPase. These data suggest that exogenous NO or NO generated by eNOS or iNOS regulates protein adduction with GSH. This could be due to a direct reaction of proteins with S-nitrosoglutathione or denitrosylation of S-nitrosylated proteins by reduced GSH. Glutathiolation of cytoskeletal and mitochondrial proteins may be a significant feature of NO bioreactivity.
DOI: 10.1042/bj20100090
2010
Cited 98 times
Role of cellular bioenergetics in smooth muscle cell proliferation induced by platelet-derived growth factor
Abnormal smooth muscle cell proliferation is a hallmark of vascular disease. Although growth factors are known to contribute to cell hyperplasia, the changes in metabolism associated with this response, particularly mitochondrial respiration, remain unclear. Given the increased energy requirements for proliferation, we hypothesized that PDGF (platelet-derived growth factor) would stimulate glycolysis and mitochondrial respiration and that this elevated bioenergetic capacity is required for smooth muscle cell hyperplasia. To test this hypothesis, cell proliferation, glycolytic flux and mitochondrial oxygen consumption were measured after treatment of primary rat aortic VSMCs (vascular smooth muscle cells) with PDGF. PDGF increased basal and maximal rates of glycolytic flux and mitochondrial oxygen consumption; enhancement of these bioenergetic pathways led to a substantial increase in the mitochondrial reserve capacity. Interventions with the PI3K (phosphoinositide 3-kinase) inhibitor LY-294002 or the glycolysis inhibitor 2-deoxy-D-glucose abrogated PDGF-stimulated proliferation and prevented augmentation of glycolysis and mitochondrial reserve capacity. Similarly, when L-glucose was substituted for D-glucose, PDGF-dependent proliferation was abolished, as were changes in glycolysis and mitochondrial respiration. Interestingly, LDH (lactate dehydrogenase) protein levels and activity were significantly increased after PDGF treatment. Moreover, substitution of L-lactate for D-glucose was sufficient to increase mitochondrial reserve capacity and cell proliferation after treatment with PDGF; these effects were inhibited by the LDH inhibitor oxamate. These results suggest that glycolysis, by providing substrates that enhance the mitochondrial reserve capacity, plays an essential role in PDGF-induced cell proliferation, underscoring the integrated metabolic response required for proliferation of VSMCs in the diseased vasculature.
DOI: 10.1152/ajpheart.00284.2007
2007
Cited 96 times
Mechanisms of acrolein-induced myocardial dysfunction: implications for environmental and endogenous aldehyde exposure
Aldehydes are ubiquitous pollutants generated during the combustion of organic materials and are present in air, water, and food. Several aldehydes are also endogenous products of lipid peroxidation and by-products of drug metabolism. Despite well-documented high reactivity of unsaturated aldehydes, little is known regarding their cardiovascular effects and their role in cardiac pathology. Accordingly, we examined the myocardial effects of the model unsaturated aldehyde acrolein. In closed-chest mice, intravenous acrolein (0.5 mg/kg) induced rapid but reversible left ventricular dilatation and dysfunction. In mouse myocytes, micromolar acrolein acutely depressed myofilament Ca(2+) responsiveness without altering catecholamine sensitivity, similar to the phenotype of stunned myocardium. Immunoblotting revealed increased acrolein-protein adducts and protein-carbonyls in both acrolein-exposed myocardium (1.8-fold increase, P < 0.002) and myocytes (6.4-fold increase, P < 0.02). Both the contractile dysfunction and adduct formation were markedly attenuated by pretreatment with the thiol donor N-acetylcysteine (5 mM). Two-dimensional gel electrophoresis and mass-assisted laser desorption/ionization time-of-flight mass spectrometry analysis revealed two groups of adducted proteins, sarcomeric/cytoskeletal proteins (cardiac alpha-actin, desmin, myosin light polypeptide 3) and energy metabolism proteins (mitochondrial creatine kinase-2, ATP synthase), indicating site-specific protein modification that was confirmed by immunohistochemical colocalization. We conclude that direct exposure to acrolein induces selective myofilament impairment, which may be, in part, related to the modification of proteins involved in myocardial contraction and energy metabolism. Myocardial dysfunction induced by acrolein and related aldehydes may be symptomatic of toxicological states associated with ambient or occupational exposures or drug toxicity. Moreover, aldehydes such as acrolein may mediate cardiac dysfunction in pathologies characterized by high-oxidative stress.
DOI: 10.1016/j.freeradbiomed.2009.06.012
2009
Cited 91 times
Methods for the determination and quantification of the reactive thiol proteome
Protein thiol modifications occur under both physiological and pathological conditions and have been shown to contribute to changes in protein structure, function, and redox signaling. The majority of protein thiol modifications occur on cysteine residues that have a low pKa; these nucleophilic proteins comprise the “reactive thiol proteome.” The most reactive members of this proteome are typically low-abundance proteins. Therefore, sensitive and quantitative methods are needed to detect and measure thiol modifications in biological samples. To accomplish this, we have standardized the usage of biotinylated and fluorophore-labeled alkylating agents, such as biotinylated iodoacetamide (IAM) and N-ethylmaleimide (NEM) and BODIPY-labeled IAM and NEM, for use in one- and two-dimensional proteomic strategies. Purified fractions of cytochrome c and glyceraldehyde-3-phosphate dehydrogenase were conjugated to a known amount of biotin or BODIPY fluorophore to create an external standard that can be run on standard SDS–PAGE gels, which allows for the quantification of protein thiols from biological samples by Western blotting or fluorescence imaging. A detailed protocol is provided for using thiol-reactive probes and making external standards for visualizing and measuring protein thiol modifications in biological samples.
DOI: 10.1016/j.bbabio.2009.11.005
2010
Cited 82 times
Regulation of vascular smooth muscle cell bioenergetic function by protein glutathiolation
Protein thiolation by glutathione is a reversible and regulated post-translational modification that is increased in response to oxidants and nitric oxide. Because many mitochondrial enzymes contain critical thiol residues, it has been hypothesized that thiolation reactions regulate cell metabolism and survival. However, it has been difficult to differentiate the biological effects due to protein thiolation from other oxidative protein modifications. In this study, we used diamide to titrate protein glutathiolation and examined its impact on glycolysis, mitochondrial function, and cell death in rat aortic smooth muscle cells. Treatment of cells with diamide increased protein glutathiolation in a concentration-dependent manner and had comparably little effect on protein–protein disulfide formation. Diamide increased mitochondrial proton leak and decreased ATP-linked mitochondrial oxygen consumption and cellular bioenergetic reserve capacity. Concentrations of diamide above 200 μM promoted acute bioenergetic failure and caused cell death, whereas lower concentrations of diamide led to a prolonged increase in glycolytic flux and were not associated with loss of cell viability. Depletion of glutathione using buthionine sulfoximine had no effect on basal protein thiolation or cellular bioenergetics but decreased diamide-induced protein glutathiolation and sensitized the cells to bioenergetic dysfunction and death. The effects of diamide on cell metabolism and viability were fully reversible upon addition of dithiothreitol. These data suggest that protein thiolation modulates key metabolic processes in both the mitochondria and cytosol.
DOI: 10.1152/ajpheart.00438.2013
2014
Cited 77 times
Cardiomyocyte <i>Ogt</i> is essential for postnatal viability
The singly coded gene O-linked-β-N-acetylglucosamine (O-GlcNAc) transferase (Ogt) resides on the X chromosome and is necessary for embryonic stem cell viability during embryogenesis. In mature cells, this enzyme catalyzes the posttranslational modification known as O-GlcNAc to various cellular proteins. Several groups, including our own, have shown that acute increases in protein O-GlcNAcylation are cardioprotective both in vitro and in vivo. Yet, little is known about how OGT affects cardiac function because total body knockout (KO) animals are not viable. Presently, we sought to establish the potential involvement of cardiomyocyte Ogt in cardiac maturation. Initially, we characterized a constitutive cardiomyocyte-specific (cm)OGT KO (c-cmOGT KO) mouse and found that only 12% of the c-cmOGT KO mice survived to weaning age (4 wk old); the surviving animals were smaller than their wild-type littermates, had dilated hearts, and showed overt signs of heart failure. Dysfunctional c-cmOGT KO hearts were more fibrotic, apoptotic, and hypertrophic. Several glycolytic genes were also upregulated; however, there were no gross changes in mitochondrial O2 consumption. Histopathology of the KO hearts indicated the potential involvement of endoplasmic reticulum stress, directing us to evaluate expression of 78-kDa glucose-regulated protein and protein disulfide isomerase, which were elevated. Additional groups of mice were subjected to inducible deletion of cmOGT, which did not produce overt dysfunction within the first couple of weeks of deletion. Yet, long-term loss (via inducible deletion) of cmOGT produced gradual and progressive cardiomyopathy. Thus, cardiomyocyte Ogt is necessary for maturation of the mammalian heart, and inducible deletion of cmOGT in the adult mouse produces progressive ventricular dysfunction.
DOI: 10.1016/j.redox.2014.12.007
2015
Cited 72 times
Autophagic regulation of smooth muscle cell biology
Autophagy regulates the metabolism, survival, and function of numerous cell types, including those comprising the cardiovascular system. In the vasculature, changes in autophagy have been documented in atherosclerotic and restenotic lesions and in hypertensive vessels. The biology of vascular smooth muscle cells appears particularly sensitive to changes in the autophagic program. Recent evidence indicates that stimuli or stressors evoked during the course of vascular disease can regulate autophagic activity, resulting in modulation of VSMC phenotype and viability. In particular, certain growth factors and cytokines, oxygen tension, and pharmacological drugs have been shown to trigger autophagy in smooth muscle cells. Importantly, each of these stimuli has a redox component, typically associated with changes in the abundance of reactive oxygen, nitrogen, or lipid species. Collective findings support the hypothesis that autophagy plays a critical role in vascular remodeling by regulating smooth muscle cell phenotype transitions and by influencing the cellular response to stress. In this graphical review, we summarize current knowledge on the role of autophagy in the biology of the smooth muscle cell in (patho)physiology.
DOI: 10.1152/physiolgenomics.00129.2012
2012
Cited 65 times
Standardized bioenergetic profiling of adult mouse cardiomyocytes
Mitochondria are at the crux of life and death and as such have become ideal targets of intervention in cardiovascular disease. Generally, current methods to measure mitochondrial dysfunction rely on working with the isolated organelle and fail to incorporate mitochondrial function in a cellular context. Extracellular flux methodology has been particularly advantageous in this respect; however, certain primary cell types, such as adult cardiac myocytes, have been difficult to standardize with this technology. Here, we describe methods for using extracellular flux (XF) analysis to measure mitochondrial bioenergetics in isolated, intact, adult mouse cardiomyocytes (ACMs). Following isolation, ACMs were seeded overnight onto laminin-coated (20 μg/ml) microplates, which resulted in high attachment efficiency. After establishing seeding density, we found that a commonly used assay medium (containing a supraphysiological concentration of pyruvate at 1 mmol/l) produced a maximal bioenergetic response. After performing a pyruvate dose-response, we determined that pyruvate titrated to 0.1 mmol/l was optimal for examining alternative substrate oxidation. Methods for measuring fatty acid oxidation were established. These methods lay the framework using XF analysis to profile metabolism of ACMs and will likely augment our ability to understand mitochondrial dysfunction in heart failure and acute myocardial ischemia. This platform could easily be extended to models of diabetes or other metabolic defects.
DOI: 10.3389/fcvm.2018.00127
2018
Cited 63 times
Metabolic Mechanisms of Exercise-Induced Cardiac Remodeling
Exercise has a myriad of physiological benefits that derive in part from its ability to improve cardiometabolic health. The periodic metabolic stress imposed by regular exercise appears fundamental in driving cardiovascular tissue adaptation. However, different types, intensities, or durations of exercise elicit different levels of metabolic stress and may promote distinct types of tissue remodeling. In this review, we discuss how exercise affects cardiac structure and function and how exercise-induced changes in metabolism regulate cardiac adaptation. Current evidence suggests that exercise typically elicits an adaptive, beneficial form of cardiac remodeling that involves cardiomyocyte growth and proliferation; however, chronic levels of extreme exercise may increase the risk for pathological cardiac remodeling or sudden cardiac death. An emerging theme underpinning acute as well as chronic cardiac adaptations to exercise is metabolic periodicity, which appears important for regulating mitochondrial quality and function, stimulating metabolism-mediated exercise gene programs and hypertrophic kinase activity, and coordinating biosynthetic pathway activity. In addition, circulating metabolites liberated during exercise trigger physiological cardiac growth. Further understanding of how exercise-mediated changes in metabolism orchestrate cell signaling and gene expression could facilitate therapeutic strategies to maximize the benefits of exercise and improve cardiac health.
DOI: 10.1042/bcj20170474
2017
Cited 57 times
Integration of flux measurements to resolve changes in anabolic and catabolic metabolism in cardiac myocytes
Although ancillary pathways of glucose metabolism are critical for synthesizing cellular building blocks and modulating stress responses, how they are regulated remains unclear. In the present study, we used radiometric glycolysis assays, [13C6]-glucose isotope tracing, and extracellular flux analysis to understand how phosphofructokinase (PFK)-mediated changes in glycolysis regulate glucose carbon partitioning into catabolic and anabolic pathways. Expression of kinase-deficient or phosphatase-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase in rat neonatal cardiomyocytes co-ordinately regulated glycolytic rate and lactate production. Nevertheless, in all groups, >40% of glucose consumed by the cells was unaccounted for via catabolism to pyruvate, which suggests entry of glucose carbons into ancillary pathways branching from metabolites formed in the preparatory phase of glycolysis. Analysis of 13C fractional enrichment patterns suggests that PFK activity regulates glucose carbon incorporation directly into the ribose and the glycerol moieties of purines and phospholipids, respectively. Pyrimidines, UDP-N-acetylhexosamine, and the fatty acyl chains of phosphatidylinositol and triglycerides showed lower 13C incorporation under conditions of high PFK activity; the isotopologue 13C enrichment pattern of each metabolite indicated limitations in mitochondria-engendered aspartate, acetyl CoA and fatty acids. Consistent with this notion, high glycolytic rate diminished mitochondrial activity and the coupling of glycolysis to glucose oxidation. These findings suggest that a major portion of intracellular glucose in cardiac myocytes is apportioned for ancillary biosynthetic reactions and that PFK co-ordinates the activities of the pentose phosphate, hexosamine biosynthetic, and glycerolipid synthesis pathways by directly modulating glycolytic intermediate entry into auxiliary glucose metabolism pathways and by indirectly regulating mitochondrial cataplerosis.
DOI: 10.1186/s12859-017-1547-6
2017
Cited 56 times
Distribution based nearest neighbor imputation for truncated high dimensional data with applications to pre-clinical and clinical metabolomics studies
High throughput metabolomics makes it possible to measure the relative abundances of numerous metabolites in biological samples, which is useful to many areas of biomedical research. However, missing values (MVs) in metabolomics datasets are common and can arise due to both technical and biological reasons. Typically, such MVs are substituted by a minimum value, which may lead to different results in downstream analyses.Here we present a modified version of the K-nearest neighbor (KNN) approach which accounts for truncation at the minimum value, i.e., KNN truncation (KNN-TN). We compare imputation results based on KNN-TN with results from other KNN approaches such as KNN based on correlation (KNN-CR) and KNN based on Euclidean distance (KNN-EU). Our approach assumes that the data follow a truncated normal distribution with the truncation point at the detection limit (LOD). The effectiveness of each approach was analyzed by the root mean square error (RMSE) measure as well as the metabolite list concordance index (MLCI) for influence on downstream statistical testing. Through extensive simulation studies and application to three real data sets, we show that KNN-TN has lower RMSE values compared to the other two KNN procedures as well as simpler imputation methods based on substituting missing values with the metabolite mean, zero values, or the LOD. MLCI values between KNN-TN and KNN-EU were roughly equivalent, and superior to the other four methods in most cases.Our findings demonstrate that KNN-TN generally has improved performance in imputing the missing values of the different datasets compared to KNN-CR and KNN-EU when there is missingness due to missing at random combined with an LOD. The results shown in this study are in the field of metabolomics but this method could be applicable with any high throughput technology which has missing due to LOD.
DOI: 10.1515/hsz-2019-0268
2019
Cited 43 times
Bioenergetics and translational metabolism: implications for genetics, physiology and precision medicine
Abstract It is now becoming clear that human metabolism is extremely plastic and varies substantially between healthy individuals. Understanding the biochemistry that underlies this physiology will enable personalized clinical interventions related to metabolism. Mitochondrial quality control and the detailed mechanisms of mitochondrial energy generation are central to understanding susceptibility to pathologies associated with aging including cancer, cardiac and neurodegenerative diseases. A precision medicine approach is also needed to evaluate the impact of exercise or caloric restriction on health. In this review, we discuss how technical advances in assessing mitochondrial genetics, cellular bioenergetics and metabolomics offer new insights into developing metabolism-based clinical tests and metabolotherapies. We discuss informatics approaches, which can define the bioenergetic-metabolite interactome and how this can help define healthy energetics. We propose that a personalized medicine approach that integrates metabolism and bioenergetics with physiologic parameters is central for understanding the pathophysiology of diseases with a metabolic etiology. New approaches that measure energetics and metabolomics from cells isolated from human blood or tissues can be of diagnostic and prognostic value to precision medicine. This is particularly significant with the development of new metabolotherapies, such as mitochondrial transplantation, which could help treat complex metabolic diseases.
DOI: 10.1093/stmcls/sxac016
2022
Cited 18 times
Metabolic Determinants of Cardiomyocyte Proliferation
The adult mammalian heart is recalcitrant to regeneration after injury, in part due to the postmitotic nature of cardiomyocytes. Accumulating evidence suggests that cardiomyocyte proliferation in fetal or neonatal mammals and in regenerative non-mammalian models depends on a conducive metabolic state. Results from numerous studies in adult hearts indicate that conditions of relatively low fatty acid oxidation, low reactive oxygen species generation, and high glycolysis are required for induction of cardiomyocyte proliferation. Glycolysis appears particularly important because it provides branchpoint metabolites for several biosynthetic pathways that are essential for synthesis of nucleotides and nucleotide sugars, amino acids, and glycerophospholipids, all of which are required for daughter cell formation. In addition, the proliferative cardiomyocyte phenotype is supported in part by relatively low oxygen tensions and through the actions of critical transcription factors, coactivators, and signaling pathways that promote a more glycolytic and proliferative cardiomyocyte phenotype, such as hypoxia inducible factor 1α (Hif1α), Yes-associated protein (Yap), and ErbB2. Interventions that inhibit glycolysis or its integrated biosynthetic pathways almost universally impair cardiomyocyte proliferative capacity. Furthermore, metabolic enzymes that augment biosynthetic capacity such as phosphoenolpyruvate carboxykinase 2 and pyruvate kinase M2 appear to be amplifiers of cardiomyocyte proliferation. Collectively, these studies suggest that acquisition of a glycolytic and biosynthetic metabolic phenotype is a sine qua non of cardiomyocyte proliferation. Further knowledge of the regulatory mechanisms that control substrate partitioning to coordinate biosynthesis with energy provision could be leveraged to prompt or augment cardiomyocyte division and to promote cardiac repair.
DOI: 10.1210/jc.2011-2963
2012
Cited 58 times
Skeletal Muscle Lipid Peroxidation and Insulin Resistance in Humans
The relationships among skeletal muscle lipid peroxidation, intramyocellular lipid content (IMCL), and insulin sensitivity were evaluated in nine insulin-sensitive (IS), 13 insulin-resistant (IR), and 10 adults with type 2 diabetes (T2DM).Insulin sensitivity was assessed by hyperinsulinemic-euglycemic clamp [glucose disposal rate (GDR)]. Lipid peroxidation was assessed by 4-hydroxynonenal (HNE)-protein adducts and general oxidative stress by protein carbonyl content. All patients were sedentary.Protein-HNE adducts were elevated 1.6-fold in T2DM compared with IS adults, whereas IR showed intermediate levels of HNE-modified proteins. Protein-HNE adducts correlated with GDR, waist circumference, and body mass index. IMCL was increased by 4.0- and 1.9-fold in T2DM and IR patients, respectively, compared with IS, and was correlated with GDR and waist circumference but not BMI. Protein carbonyls were not different among groups and did not correlate with any of the measured variables. Correlations were detected between IMCL and protein-HNE.Our data show for the first time that skeletal muscle protein-HNE adducts are related to the severity of insulin resistance in sedentary adults. These results suggest that muscle lipid peroxidation could be involved in the development of insulin resistance.
DOI: 10.1002/stem.1325
2013
Cited 55 times
Protein <i>O</i>-GlcNAcylation Is a Novel Cytoprotective Signal in Cardiac Stem Cells
Clinical trials demonstrate the regenerative potential of cardiac stem cell (CSC) therapy in the postinfarcted heart. Despite these encouraging preliminary clinical findings, the basic biology of these cells remains largely unexplored. The principal requirement for cell transplantation is to effectively prime them for survival within the unfavorable environment of the infarcted myocardium. In the adult mammalian heart, the β-O-linkage of N-acetylglucosamine (i.e., O-GlcNAc) to proteins is a unique post-translational modification that confers cardioprotection from various otherwise lethal stressors. It is not known whether this signaling system exists in CSCs. In this study, we demonstrate that protein O-GlcNAcylation is an inducible stress response in adult murine Sca-1(+) /lin(-) CSCs and exerts an essential prosurvival role. Posthypoxic CSCs responded by time-dependently increasing protein O-GlcNAcylation upon reoxygenation. We used pharmacological interventions for loss- and gain-of-function, that is, enzymatic inhibition of O-GlcNAc transferase (OGT) (adds the O-GlcNAc modification to proteins) by TT04, or inhibition of OGA (removes O-GlcNAc) by thiamet-G (ThG). Reduction in the O-GlcNAc signal (via TT04, or OGT gene deletion using Cre-mediated recombination) significantly sensitized CSCs to posthypoxic injury, whereas augmenting O-GlcNAc levels (via ThG) enhanced cell survival. Diminished O-GlcNAc levels render CSCs more susceptible to the onset of posthypoxic apoptotic processes via elevated poly(ADP-ribose) polymerase cleavage due to enhanced caspase-3/7 activation, whereas promoting O-GlcNAcylation can serve as a pre-emptive antiapoptotic signal regulating the survival of CSCs. Thus, we report the primary demonstration of protein O-GlcNAcylation as an important prosurvival signal in CSCs, which could enhance CSC survival prior to in vivo autologous transfer.
DOI: 10.1002/stem.2047
2015
Cited 48 times
Glutamine Regulates Cardiac Progenitor Cell Metabolism and Proliferation
Abstract Autologous transplantation of cardiac progenitor cells (CPCs) alleviates myocardial dysfunction in the damaged heart; however, the mechanisms that contribute to their reparative qualities remain poorly understood. In this study, we examined CPC metabolism to elucidate the metabolic pathways that regulate their proliferative capacity. In complete growth medium, undifferentiated CPCs isolated from adult mouse heart proliferated rapidly (Td = 13.8 hours). CPCs expressed the Glut1 transporter and their glycolytic rate was increased by high extracellular glucose (Glc) concentration, in the absence of insulin. Although high Glc concentrations did not stimulate proliferation, glutamine (Gln) increased CPC doubling time and promoted survival under conditions of oxidative stress. In comparison with Glc, pyruvate (Pyr) or BSA-palmitate, Gln, when provided as the sole metabolic substrate, increased ATP-linked and uncoupled respiration. Although fatty acids were not used as respiratory substrates when present as a sole carbon source, Gln-induced respiration was doubled in the presence of BSA-palmitate, suggesting that Gln stimulates fatty acid oxidation. Additionally, Gln promoted rapid phosphorylation of the mTORC1 substrate, p70S6k, as well as retinoblastoma protein, followed by induction of cyclin D1 and cdk4. Inhibition of either mTORC1 or glutaminolysis was sufficient to diminish CPC proliferation, and provision of cell permeable α-ketoglutarate in the absence of Gln increased both respiration and cell proliferation, indicating a key role of Gln anaplerosis in cell growth. These findings suggest that Gln, by enhancing mitochondrial function and stimulating mTORC1, increases CPC proliferation, and that interventions to increase Gln uptake or oxidation may improve CPC therapy. Stem Cells 2015;33:2613—2627
DOI: 10.1042/bj20131608
2014
Cited 48 times
Bioenergetic differences between MCF-7 and T47D breast cancer cells and their regulation by oestradiol and tamoxifen
Oestrogen receptor α (ERα+) breast tumours rely on mitochondria (mt) to generate ATP. The goal of the present study was to determine how oestradiol (E2) and 4-hydroxytamoxifen (4-OHT) affect cellular bioenergetic function in MCF-7 and T47D ERα+ breast cancer cells in serum-replete compared with dextran-coated charcoal (DCC)-stripped foetal bovine serum (FBS)-containing medium (‘serum-starved’). Serum-starvation reduced oxygen consumption rate (OCR), extracellular acidification rate (ECAR), ATP-linked OCR and maximum mt capacity, reflecting lower ATP demand and mt respiration. Cellular respiratory stateapparent was unchanged by serum deprivation. 4-OHT reduced OCR independent of serum status. Despite having a higher mt DNA/nuclear DNA ratio than MCF-7 cells, T47D cells have a lower OCR and ATP levels and higher proton leak. T47D express higher nuclear respiratory factor-1 (NRF-1) and NRF-1-regulated, nuclear-encoded mitochondrial transcription factor TFAM and cytochrome c, but lower levels of cytochrome c oxidase, subunit IV, isoform 1 (COX4, COX4I1). Mitochondrial reserve capacity, reflecting tolerance to cellular stress, was higher in serum-starved T47D cells and was increased by 4-OHT, but was decreased by 4-OHT in MCF-7 cells. These data demonstrate critical differences in cellular energetics and responses to 4-OHT in these two ERα+ cell lines, likely reflecting cancer cell avoidance of apoptosis.
DOI: 10.1042/bj20141018
2015
Cited 42 times
High glucose induces mitochondrial dysfunction independently of protein O-GlcNAcylation
Diabetes is characterized by hyperglycaemia and perturbations in intermediary metabolism. In particular, diabetes can augment flux through accessory pathways of glucose metabolism, such as the hexosamine biosynthetic pathway (HBP), which produces the sugar donor for the β-O-linked-N-acetylglucosamine (O-GlcNAc) post-translational modification of proteins. Diabetes also promotes mitochondrial dysfunction. Nevertheless, the relationships among diabetes, hyperglycaemia, mitochondrial dysfunction and O-GlcNAc modifications remain unclear. In the present study, we tested whether high-glucose-induced increases in O-GlcNAc modifications directly regulate mitochondrial function in isolated cardiomyocytes. Augmentation of O-GlcNAcylation with high glucose (33 mM) was associated with diminished basal and maximal cardiomyocyte respiration, a decreased mitochondrial reserve capacity and lower Complex II-dependent respiration (P<0.05); however, pharmacological or genetic modulation of O-GlcNAc modifications under normal or high glucose conditions showed few significant effects on mitochondrial respiration, suggesting that O-GlcNAc does not play a major role in regulating cardiomyocyte mitochondrial function. Furthermore, an osmotic control recapitulated high-glucose-induced changes to mitochondrial metabolism (P<0.05) without increasing O-GlcNAcylation. Thus, increased O-GlcNAcylation is neither sufficient nor necessary for high-glucose-induced suppression of mitochondrial metabolism in isolated cardiomyocytes.
DOI: 10.1172/jci.insight.98441
2018
Cited 41 times
TAK1 regulates skeletal muscle mass and mitochondrial function
Skeletal muscle mass is regulated by a complex array of signaling pathways. TGF-β-activated kinase 1 (TAK1) is an important signaling protein, which regulates context-dependent activation of multiple intracellular pathways. However, the role of TAK1 in the regulation of skeletal muscle mass remains unknown. Here, we report that inducible inactivation of TAK1 causes severe muscle wasting, leading to kyphosis, in both young and adult mice.. Inactivation of TAK1 inhibits protein synthesis and induces proteolysis, potentially through upregulating the activity of the ubiquitin-proteasome system and autophagy. Phosphorylation and enzymatic activity of AMPK are increased, whereas levels of phosphorylated mTOR and p38 MAPK are diminished upon inducible inactivation of TAK1 in skeletal muscle. In addition, targeted inactivation of TAK1 leads to the accumulation of dysfunctional mitochondria and oxidative stress in skeletal muscle of adult mice. Inhibition of TAK1 does not attenuate denervation-induced muscle wasting in adult mice. Finally, TAK1 activity is highly upregulated during overload-induced skeletal muscle growth, and inactivation of TAK1 prevents myofiber hypertrophy in response to functional overload. Overall, our study demonstrates that TAK1 is a key regulator of skeletal muscle mass and oxidative metabolism.
DOI: 10.1007/s00395-018-0703-0
2018
Cited 41 times
Cardiac mesenchymal cells from diabetic mice are ineffective for cell therapy-mediated myocardial repair
Although cell therapy improves cardiac function after myocardial infarction, highly variable results and limited understanding of the underlying mechanisms preclude its clinical translation. Because many heart failure patients are diabetic, we examined how diabetic conditions affect the characteristics of cardiac mesenchymal cells (CMC) and their ability to promote myocardial repair in mice. To examine how diabetes affects CMC function, we isolated CMCs from non-diabetic C57BL/6J (CMCWT) or diabetic B6.BKS(D)-Leprdb/J (CMCdb/db) mice. When CMCs were grown in 17.5 mM glucose, CMCdb/db cells showed > twofold higher glycolytic activity and a threefold higher expression of Pfkfb3 compared with CMCWT cells; however, culture of CMCdb/db cells in 5.5 mM glucose led to metabolic remodeling characterized by normalization of metabolism, a higher NAD+/NADH ratio, and a sixfold upregulation of Sirt1. These changes were associated with altered extracellular vesicle miRNA content as well as proliferation and cytotoxicity parameters comparable to CMCWT cells. To test whether this metabolic improvement of CMCdb/db cells renders them suitable for cell therapy, we cultured CMCWT or CMCdb/db cells in 5.5 mM glucose and then injected them into infarcted hearts of non-diabetic mice (CMCWT, n = 17; CMCdb/db, n = 13; Veh, n = 14). Hemodynamic measurements performed 35 days after transplantation showed that, despite normalization of their properties in vitro, and unlike CMCWT cells, CMCdb/db cells did not improve load-dependent and -independent parameters of left ventricular function. These results suggest that diabetes adversely affects the reparative capacity of CMCs and that modulating CMC characteristics via culture in lower glucose does not render them efficacious for cell therapy.
DOI: 10.1152/ajpheart.00881.2020
2021
Cited 27 times
Fine particulate matter (PM<sub>2.5</sub>) inhalation-induced alterations in the plasma lipidome as promoters of vascular inflammation and insulin resistance
Fine particulate matter (PM2.5) air pollution exposure increases the risk of developing cardiovascular disease (CVD). Although the precise mechanisms by which air pollution exposure increases CVD risk remain uncertain, research indicates that PM2.5-induced endothelial dysfunction contributes to CVD risk. Previous studies demonstrate that concentrated ambient PM2.5 (CAP) exposure induces vascular inflammation and impairs insulin and vascular endothelial growth factor (VEGF) signaling dependent on pulmonary oxidative stress. To assess whether CAP exposure induces these vascular effects via plasmatic factors, we incubated aortas from naïve mice with plasma isolated from mice exposed to HEPA-filtered air or CAP (9 days) and examined vascular inflammation and insulin and VEGF signaling. We found that treatment of naïve aortas with plasma from CAP-exposed mice activates NF-κBα and induces insulin and VEGF resistance, indicating transmission by plasmatic factor(s). To identify putative factors, we exposed lung-specific ecSOD-transgenic (ecSOD-Tg) mice and wild-type (WT) littermates to CAP at concentrations of either ∼60 µg/m3 (CAP60) or ∼100 µg/m3 (CAP100) and measured the abundance of plasma metabolites by mass spectrometry. In WT mice, both CAP concentrations increased levels of fatty acids such as palmitate, myristate, and palmitoleate and decreased numerous phospholipid species; however, these CAP-induced changes in the plasma lipidome were prevented in ecSOD-Tg mice. Consistent with the literature, we found that fatty acids such as palmitate are sufficient to promote endothelial inflammation. Collectively, our findings suggest that PM2.5 exposure, by inducing pulmonary oxidative stress, promotes unique lipidomic changes characterized by high levels of circulating fatty acids, which are sufficient to trigger vascular pathology.NEW & NOTEWORTHY We found that circulating plasma constituents are responsible for air pollution-induced vascular pathologies. Inhalation of fine particulate matter (≤PM2.5) promotes a unique form of dyslipidemia that manifests in a manner dependent upon pulmonary oxidative stress. The air pollution-engendered dyslipidemic phenotype is characterized by elevated free fatty acid species and diminished phospholipid species, which could contribute to vascular inflammation and loss of insulin sensitivity.
DOI: 10.1161/circulationaha.121.057879
2022
Cited 15 times
Glutaminolysis is Essential for Myofibroblast Persistence and In Vivo Targeting Reverses Fibrosis and Cardiac Dysfunction in Heart Failure
HomeCirculationVol. 145, No. 21Glutaminolysis is Essential for Myofibroblast Persistence and In Vivo Targeting Reverses Fibrosis and Cardiac Dysfunction in Heart Failure Free AccessLetterPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessLetterPDF/EPUBGlutaminolysis is Essential for Myofibroblast Persistence and In Vivo Targeting Reverses Fibrosis and Cardiac Dysfunction in Heart Failure Andrew A. Gibb, PhD, Emma K. Murray, PhD, Anh T. Huynh, BS, Ryan B. Gaspar, Tori L. Ploesch, Ken Bedi, BS, Alyssa A. Lombardi, MD, PhD, Pawel K. Lorkiewicz, PhD, Rajika Roy, PhD, Zolt Arany, MD, PhD, Daniel P. Kelly, MD, Kenneth B. Margulies, MD, Bradford G. Hill, PhD and John W. Elrod, PhD Andrew A. GibbAndrew A. Gibb https://orcid.org/0000-0003-2770-430X Center for Translational Medicine, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (A.A.G., E.K.M., A.T.H., R.B.G., T.L.P., A.A.L., R.R., J.W.E.). , Emma K. MurrayEmma K. Murray https://orcid.org/0000-0001-5871-0564 Center for Translational Medicine, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (A.A.G., E.K.M., A.T.H., R.B.G., T.L.P., A.A.L., R.R., J.W.E.). , Anh T. HuynhAnh T. Huynh Center for Translational Medicine, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (A.A.G., E.K.M., A.T.H., R.B.G., T.L.P., A.A.L., R.R., J.W.E.). , Ryan B. GasparRyan B. Gaspar https://orcid.org/0000-0002-7922-3893 , Tori L. PloeschTori L. Ploesch https://orcid.org/0000-0001-5089-3255 , Ken BediKen Bedi Cardiovascular Institute and Cardiovascular Medicine Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (K.B., Z.A., D.P.K., K.B.M.). , Alyssa A. LombardiAlyssa A. Lombardi Center for Translational Medicine, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (A.A.G., E.K.M., A.T.H., R.B.G., T.L.P., A.A.L., R.R., J.W.E.). , Pawel K. LorkiewiczPawel K. Lorkiewicz https://orcid.org/0000-0002-1755-9826 Department of Chemistry (P.K.L.), University of Louisville, KY. , Rajika RoyRajika Roy Center for Translational Medicine, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (A.A.G., E.K.M., A.T.H., R.B.G., T.L.P., A.A.L., R.R., J.W.E.). , Zolt AranyZolt Arany https://orcid.org/0000-0003-1368-2453 Cardiovascular Institute and Cardiovascular Medicine Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (K.B., Z.A., D.P.K., K.B.M.). , Daniel P. KellyDaniel P. Kelly https://orcid.org/0000-0002-3811-9491 Cardiovascular Institute and Cardiovascular Medicine Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (K.B., Z.A., D.P.K., K.B.M.). , Kenneth B. MarguliesKenneth B. Margulies https://orcid.org/0000-0002-8093-4465 Cardiovascular Institute and Cardiovascular Medicine Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (K.B., Z.A., D.P.K., K.B.M.). , Bradford G. HillBradford G. Hill Division of Environmental Medicine (B.G.H.), University of Louisville, KY. and John W. ElrodJohn W. Elrod Correspondence to: John W. Elrod, PhD, Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, MERB 949, Philadelphia, PA 19140. Email E-mail Address: [email protected] https://orcid.org/0000-0003-3925-2224 Center for Translational Medicine, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (A.A.G., E.K.M., A.T.H., R.B.G., T.L.P., A.A.L., R.R., J.W.E.). Originally published23 May 2022https://doi.org/10.1161/CIRCULATIONAHA.121.057879Circulation. 2022;145:1625–1628After injury, cardiac fibroblasts (CFs) differentiate into highly specialized myofibroblasts, essential for maintaining myocardial structural integrity by contracting surrounding tissue and remodeling the extracellular matrix network.1 It was recently reported that withdrawal of stress stimuli is sufficient for some myofibroblasts to revert to a quiescent phenotype,2 suggesting the potential for myofibroblast reversion by targeting the molecular pathways maintaining the fibrotic phenotype. Metabolic reprogramming has become appreciated as a primary trigger of cell fate transition, directing metabolic intermediates into pathways necessary for bioenergetics, anabolism, and the synthesis of cofactors regulating epigenetic modifiers and downstream gene expression. We recently reported that glutaminolysis (glutamine→glutamate) was linked to histone demethylation mediating myofibroblast formation,3 implicating metabolic remodeling as a critical feature of the differentiation program. However, the greatest therapeutic potential of targeting the myofibroblast phenotype lies in reversing fibrosis by dedifferentiating activated myofibroblasts after injury under chronic stress stimuli.To determine whether glutaminolysis inhibition is sufficient to revert myofibroblasts to a quiescent phenotype and reverse tissue fibrosis, we targeted Gls1 (glutaminase-1), the replace with committed step in of glutaminolysis, following fibroblast activation in a murine model of pressure-overload heart failure (HF). Gls1fl/fl mice4 were crossed with a Periostin inducible-Cre mouse line, restricting Cre recombination to activated fibroblasts (PostniCre.)2 Because periostin is only expressed after fibroblast activation and the in vivo half-life of GLS1 is ~5.1 days,5 this strategy allows for deletion of Gls1 exclusively after myofibroblast differentiation. Given the difficulty in accurately identifying fibroblasts by surface antigens, Gls1fl/fl×PostniCre mice were subsequently crossed to a Cre-dependent reporter mouse line (tdTomato) to permit isolation and assessment of any activated fibroblasts previously or currently expressing periostin (Figure, A). Mice were fed tamoxifen chow (tamox; 40 mg/kg/d) 1 day after transverse aortic constriction (TAC) and for the remainder of the study. Maintenance on tamox allowed for tdTomato labeling and Gls1 deletion in all CFs activated during pressure overload. Cardiac function was monitored before and every 2 weeks after TAC (Figure, B). All animal work was performed in accordance with Institutional Animal Care and Use Committee approval by Temple University and followed American Association for Accreditation of Laboratory Animal Care guidelines. The data that support the findings of this study are available from the corresponding author upon reasonable request.Download figureDownload PowerPointFigure. Inhibition of glutaminolysis in activated myofibroblasts reverses cardiac functional decline and fibrosis after murine pressure overload and reverts myofibroblasts isolated from nonischemic cardiomyopathy patients to a less fibrotic phenotype. A, Gls1fl/fl mice were crossed with mice expressing a tamoxifen (tamox)–inducible, fibroblast-specific Cre recombinase (PostniCre), allowing for Gls1 (glutaminase-1)–specific deletion in activated myofibroblasts, after differentiation. In addition, these mice were crossed to a Cre-dependent reporter line to allow for isolation and assessment of the activated and Gls1-deleted fibroblast population. B, Schematic of experimental timeline. Mice were fed tamox chow 1 day after transverse aortic constriction (TAC) and maintained on tamox chow for the duration of the study. Echocardiography was assessed at baseline and every 2 weeks after surgery. C, Western blot of tdTomato+ cells collected through fluorescence-activated cell sorting to confirm GLS1 deletion (KGA, full length; GAC, splice variant isoform). D, Aortic pressures 1 week after TAC. E, Total TdTomato+ cells from hearts 16 weeks after TAC. F, Ratio of heart weight to tibia length (HW/TL) 16 weeks after TAC. G, Short-axis M-mode echo measurements of percent fractional shortening (FS%). Long-axis B-mode echo measurements of (H) end diastolic volume (EDV), (I) end systolic volume (ESV), and (J) percent ejection fraction (EF%) 16 weeks after TAC. K, Long-axis B-mode measurements of radial myocardial strain rates using speckle tracking echocardiography. L, Inverse strain rates indicative of the rate of myocardial relaxation. M, Representative images of hearts stained with picrosirius red to visualize and quantify tissue fibrosis after 16 weeks of TAC. Scale bar, 250 μm. N, Quantification of images in (M) as the percent of red stained area to total tissue area. O, Hydroxyproline content in left ventricular (LV) tissue samples. P, Schematic of cell acquisition, isolation, and experimental timeline to assess GLS1 inhibition on myofibroblast reversion under sustained stress stimuli (transforming growth factor–β [TGFβ]) in cardiac fibroblasts obtained from patients with nonischemic cardiomyopathy. Q and T, Representative images of cardiac fibroblasts chronically treated ± TGFβ for 48 hours. After the first 24-hour period, once myofibroblast formation had occurred, GLS1 inhibitor CB-839 (10 μM) was added and stained for α–smooth muscle actin (αSMA; red) and nuclear stain (DAPI; blue). Scale bar, 40 μm. R and U, Quantification of images in Q and T as the percent of cells expressing αSMA to the total number of cells (minimum of 80 cells quantified per replicate). S and V, Relative fold change in expression of myofibroblast genes (ACTA2, FN1, POSTN, PDGFRA, COL1A1, and COL3A1). RPS13 was used as the loading control. Data shown as mean ± SEM, n=2 to 15 per group, as indicated. For statistical considerations, unpaired t tests (D, E, and H–L), 1-way analyses of variance with the Tukey post hoc test (R, S, U, and V), 2-way analyses of variance with the Tukey post hoc test (F, N, and O), or a 2-way mixed-effects model analysis of variance with Sidak post hoc test for multiple comparisons (G) was used (*P<0.05, **P<0.01, ***P<0.001, ****P<0.0001). qPCR indicates quantitative polymerase chain reaction.Western blot analysis of tdTomato+ CFs isolated by fluorescence-activated cell sorting confirmed Gls1 deletion after TAC (Figure, C). Aortic pressures obtained 1 week after TAC indicated similar pressure gradients between control (PostniCre) and experimental (Gls1fl/fl×PostniCre) mice (Figure, D). Determination of the number of CFs that had been activated after TAC, ~3% of all gated cells, revealed no significant difference between groups, confirming we were temporally deleting Gls1 after fibroblast activation and differentiation (Figure, E). We did not observe any difference in cardiac hypertrophy (Figure, F), but we did observe preserved left ventricular fractional shortening from 10 to 16 weeks after TAC in Gls1fl/fl×PostniCre mice (Figure, G). To further examine cardiac contractility and relaxation, B-mode speckle-tracking echocardiography was performed. Ejection fraction rebounded in Gls1fl/fl×PostniCre mice 16 weeks after TAC (Figure, H–J). We also found left ventricular radial strain rates in Gls1fl/fl×PostniCre mice to be significantly greater than in controls, indicating increased systolic function (Figure, K). We also saw enhanced inverse radial strain rates, a measure of the rate of left ventricular relaxation or diastolic function (Figure, L), an important finding because diastolic dysfunction highly correlates with tissue fibrosis. Assessment of collagen deposition (% fibrosis) revealed a significant increase in left ventricular fibrosis in control mice 16 weeks after TAC, which was prevented by targeting Gls1 in activated myofibroblasts (Figure, M–O). It is important to note, collagen deposition was similar between groups at an earlier time point (6 weeks after TAC; Figure, O), confirming our hypothesis of fibrotic regression after loss of glutaminolysis in activated myofibroblasts. These in vivo results indicate the therapeutic potential of targeting glutaminolysis in activated myofibroblasts to reverse cardiac fibrosis and mitigate functional decline in HF.To further determine the translational potential of targeting glutaminolysis for myofibroblast reversion, we isolated CFs from heart tissue of patients with nonischemic cardiomyopathy and maintained these cells under disease conditions (transforming growth factor–β stimulation for 48 hours), treating with a GLS1 inhibitor (CB-839; 10 μM) at the 24-hour time point (Figure, P). Research use of human heart tissues in this study was approved by the University of Pennsylvania institutional review board with written informed consent obtained from each of the heart transplant recipients providing explanted heart tissue for this research. Analysis of α–smooth muscle actin+ cells (% αSMA+, myofibroblast number) and activation state of the fibrotic gene program by quantitative polymerase chain reaction revealed pharmacologic inhibition of glutaminolysis in CFs isolated from patients with HF was sufficient to decrease myofibroblast number and revert the gene program to a less fibrotic phenotype (Figure, Q–V). These results provide direct evidence for therapeutically targeting glutaminolysis in patients with HF to positively alter the fibroblast population and curb active fibrosis.Our results identify glutaminolysis as a critical mediator of myofibroblast persistence in a murine model of HF and in CFs isolated directly from failing human hearts. This could be attributable to the essential role of glutamine as a carbon source for bioenergetics, anabolism, collagen biosynthesis, and epigenetic regulation of gene expression, all of which contribute to myofibroblast differentiation and the fibrotic phenotype. Small molecule inhibition of GLS1 is in phase 2 cancer clinical trials, which if found to be safe, could be repurposed rapidly for the treatment of fibrotic disease.Article InformationAcknowledgmentsThe authors thank Trevor Tierney and Alycia Hildebrand for management of the laboratory and mutant mouse colony. Author contributions are as follows: methodology: Drs Gibb and Elrod; data collection: Drs Gibb and Murray, A.T. Huynh, R.B. Gaspar, T.L. Ploesch, K. Bedi, and Drs Lombardi, Lorkiewicz, and Elrod; formal analysis: Drs Gibb, Lorkiewicz, Hill, and Elrod; resources: Drs Gibb, Elrod, Arany, Kelly, Margulies, and Hill; and manuscript writing: Drs Gibb and Elrod. All authors gave approval for the final version of the article.Sources of FundingThis work was supported in part by grants from the National Institutes of Health to Drs Elrod (grants R01HL123966, R01HL136954, R01HL142271, and P01HL134608), Gibb (grant F32HL145914), Murray (grant F31HL143913), Arany (grant R01HL152446), Kelly (grant R01HL058493), Margulies (grants R01HL149891 and R01HL105993), and Hill (grant R01HL130174) and from the American Heart Association to Dr Elrod (grant 20EIA35320226).Nonstandard Abbreviations and AcronymsCFcardiac fibroblastGLS1glutaminase-1HFheart failureTACtransverse aortic constrictionDisclosures Dr Kelly is a scientific consultant for Pfizer Inc and Amgen Inc. Dr Margulies is a consultant for Bristol Myers Squibb and has received research funding from Amgen, Sanofi-Aventis USA, and Merck Sharp & Dohme. Dr Elrod is a scientific consultant for Mitobridge Inc and Janssen and receives research funding from Mitobridge Inc.FootnotesCirculation is available at www.ahajournals.org/journal/circFor Sources of Funding and Disclosures, see page 1628.Correspondence to: John W. Elrod, PhD, Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, MERB 949, Philadelphia, PA 19140. Email [email protected]eduReferences1. Gibb AA, Lazaropoulos MP, Elrod JW. Myofibroblasts and fibrosis: mitochondrial and metabolic control of cellular differentiation.Circ Res. 2020; 127:427–447. doi: 10.1161/CIRCRESAHA.120.316958LinkGoogle Scholar2. Kanisicak O, Khalil H, Ivey MJ, et al. Genetic lineage tracing defines myofibroblast origin and function in the injured heart.Nat Commun. 2016; 7:12260. doi: 10.1038/ncomms12260CrossrefMedlineGoogle Scholar3. Lombardi AA, Gibb AA, Arif E, et al. Mitochondrial calcium exchange links metabolism with the epigenome to control cellular differentiation.Nat Commun. 2019; 10:4509. doi: 10.1038/s41467-019-12103-xCrossrefMedlineGoogle Scholar4. Mingote S, Masson J, Gellman C, et al. Genetic pharmacotherapy as an early CNS drug development strategy: testing glutaminase inhibition for schizophrenia treatment in adult mice.Front Syst Neurosci. 2015; 9:165. doi: 10.3389/fnsys.2015.00165MedlineGoogle Scholar5. Tong J, Harrison G, Curthoys NP. The effect of metabolic acidosis on the synthesis and turnover of rat renal phosphate-dependent glutaminase.Biochem J. 1986; 233:139–144. doi: 10.1042/bj2330139CrossrefMedlineGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetails May 24, 2022Vol 145, Issue 21 Advertisement Article InformationMetrics © 2022 American Heart Association, Inc.https://doi.org/10.1161/CIRCULATIONAHA.121.057879PMID: 35605036 Originally publishedMay 23, 2022 Keywordsmetabolismheart failureglutaminemyofibroblastsfibrosisPDF download Advertisement SubjectsFibrosisHeart FailureMetabolismRemodeling
DOI: 10.1093/toxsci/kfw045
2016
Cited 38 times
Vinyl Chloride Metabolites Potentiate Inflammatory Liver Injury Caused by LPS in Mice
Vinyl chloride (VC) is a ubiquitous environmental contaminant for which human risk is incompletely understood. We have previously reported that high occupational exposure to VC directly caused liver damage in humans. However, whether VC may also potentiate liver injury from other causes is not known. C57Bl/6J mice were administered chloroethanol (CE), a major metabolite of VC, and lipopolysaccharide (LPS) 24 h after CE. Samples were harvested for determination of liver damage, inflammation, and changes in carbohydrate and lipid metabolism. In mice, CE exposure alone caused no detectable liver damage. LPS exposure caused inflammatory liver damage, oxidative stress, lipid accumulation, and glycogen depletion; the effect of all of these variables was potentiated by CE pre-exposure. In vitro experiments suggest that VC metabolite chloroacetaldehyde (CAA) directly damages mitochondria, which may explain the sensitization effect observed in vivo Moreover, co-exposure of cells to CAA and TNFα caused increased cell death, supporting the hypothesis of sensitization by VC metabolites. Taken together, these data demonstrate that exposure to VC/metabolites at levels that are not overtly hepatotoxic can potentiate liver injury caused by another hepatotoxicant. This serves as proof-of-concept that VC hepatotoxicity may be modified by an additional metabolic stress such as endotoxemia, which commonly occurs in acute (eg, sepsis) and chronic (eg, NAFLD) diseases.
DOI: 10.1161/circresaha.114.305518
2015
Cited 36 times
Genetic Deficiency of Glutathione <i>S</i> -Transferase P Increases Myocardial Sensitivity to Ischemia–Reperfusion Injury
Myocardial ischemia-reperfusion (I/R) results in the generation of oxygen-derived free radicals and the accumulation of lipid peroxidation-derived unsaturated aldehydes. However, the contribution of aldehydes to myocardial I/R injury has not been assessed.We tested the hypothesis that removal of aldehydes by glutathione S-transferase P (GSTP) diminishes I/R injury.In adult male C57BL/6 mouse hearts, Gstp1/2 was the most abundant GST transcript followed by Gsta4 and Gstm4.1, and GSTP activity was a significant fraction of the total GST activity. mGstp1/2 deletion reduced total GST activity, but no compensatory increase in GSTA and GSTM or major antioxidant enzymes was observed. Genetic deficiency of GSTP did not alter cardiac function, but in comparison with hearts from wild-type mice, the hearts isolated from GSTP-null mice were more sensitive to I/R injury. Disruption of the GSTP gene also increased infarct size after coronary occlusion in situ. Ischemia significantly increased acrolein in hearts, and GSTP deficiency induced significant deficits in the metabolism of the unsaturated aldehyde, acrolein, but not in the metabolism of 4-hydroxy-trans-2-nonenal or trans-2-hexanal; on ischemia, the GSTP-null hearts accumulated more acrolein-modified proteins than wild-type hearts. GSTP deficiency did not affect I/R-induced free radical generation, c-Jun N-terminal kinase activation, or depletion of reduced glutathione. Acrolein exposure induced a hyperpolarizing shift in INa, and acrolein-induced cell death was delayed by SN-6, a Na(+)/Ca(++) exchange inhibitor. Cardiomyocytes isolated from GSTP-null hearts were more sensitive than wild-type myocytes to acrolein-induced protein crosslinking and cell death.GSTP protects the heart from I/R injury by facilitating the detoxification of cytotoxic aldehydes, such as acrolein.
DOI: 10.1074/jbc.m116.722496
2016
Cited 36 times
Type 2 Diabetes Dysregulates Glucose Metabolism in Cardiac Progenitor Cells
Type 2 diabetes is associated with increased mortality and progression to heart failure. Recent studies suggest that diabetes also impairs reparative responses after cell therapy. In this study, we examined potential mechanisms by which diabetes affects cardiac progenitor cells (CPCs). CPCs isolated from the diabetic heart showed diminished proliferation, a propensity for cell death, and a pro-adipogenic phenotype. The diabetic CPCs were insulin-resistant, and they showed higher energetic reliance on glycolysis, which was associated with up-regulation of the pro-glycolytic enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3). In WT CPCs, expression of a mutant form of PFKFB, which mimics PFKFB3 activity and increases glycolytic rate, was sufficient to phenocopy the mitochondrial and proliferative deficiencies found in diabetic cells. Consistent with activation of phosphofructokinase in diabetic cells, stable isotope carbon tracing in diabetic CPCs showed dysregulation of the pentose phosphate and glycero(phospho)lipid synthesis pathways. We describe diabetes-induced dysregulation of carbon partitioning using stable isotope metabolomics-based coupling quotients, which relate relative flux values between metabolic pathways. These findings suggest that diabetes causes an imbalance in glucose carbon allocation by uncoupling biosynthetic pathway activity, which could diminish the efficacy of CPCs for myocardial repair.
DOI: 10.1016/j.yexcr.2016.08.006
2016
Cited 36 times
Nuclear respiratory factor-1 and bioenergetics in tamoxifen-resistant breast cancer cells
Acquired tamoxifen (TAM) resistance is a significant clinical problem in treating patients with estrogen receptor α (ERα)+ breast cancer. We reported that ERα increases nuclear respiratory factor-1 (NRF-1), which regulates nuclear-encoded mitochondrial gene transcription, in MCF-7 breast cancer cells and NRF-1 knockdown stimulates apoptosis. Whether NRF-1 and target gene expression is altered in endocrine resistant breast cancer cells is unknown. We measured NRF-1and metabolic features in a cell model of progressive TAM-resistance. NRF-1 and its target mitochondrial transcription factor A (TFAM) were higher in TAM-resistant LCC2 and LCC9 cells than TAM-sensitive MCF-7 cells. Using extracellular flux assays we observed that LCC1, LCC2, and LCC9 cells showed similar oxygen consumption rate (OCR), but lower mitochondrial reserve capacity which was correlated with lower Succinate Dehydrogenase Complex, Subunit B in LCC1 and LCC2 cells. Complex III activity was lower in LCC9 than MCF-7 cells. LCC1, LCC2, and LCC9 cells had higher basal extracellular acidification (ECAR), indicating higher aerobic glycolysis, relative to MCF-7 cells. Mitochondrial bioenergetic responses to estradiol and 4-hydroxytamoxifen were reduced in the endocrine-resistant cells compared to MCF-7 cells. These results suggest the acquisition of altered metabolic phenotypes in response to long term antiestrogen treatment may increase vulnerability to metabolic stress.
DOI: 10.2337/db15-1689
2016
Cited 32 times
CCR7 Maintains Nonresolving Lymph Node and Adipose Inflammation in Obesity
Accumulation of immune cells in adipose tissue promotes insulin resistance in obesity. Although innate and adaptive immune cells contribute to adipose inflammation, the processes that sustain these interactions are incompletely understood. Here we show that obesity promotes the accumulation of CD11c(+) adipose tissue immune cells that express C-C chemokine receptor 7 (CCR7) in mice and humans, and that CCR7 contributes to chronic inflammation and insulin resistance. We identified that CCR7(+) macrophages and dendritic cells accumulate in adipose tissue in close proximity to lymph nodes (LNs) (i.e., perinodal) and visceral adipose. Consistent with the role of CCR7 in regulating the migration of immune cells to LNs, obesity promoted the accumulation of CD11c(+) cells in LNs, which was prevented by global or hematopoietic deficiency of Ccr7 Obese Ccr7(-/-) mice had reduced accumulation of CD8(+) T cells, B cells, and macrophages in adipose tissue, which was associated with reduced inflammatory signaling. This reduction in maladaptive inflammation translated to increased insulin signaling and improved glucose tolerance in obesity. Therapeutic administration of an anti-CCR7 antibody phenocopied the effects of genetic Ccr7 deficiency in mice with established obesity. These results suggest that CCR7 plays a causal role in maintaining innate and adaptive immunity in obesity.
DOI: 10.1002/reg2.95
2018
Cited 32 times
High throughput measurement of metabolism in planarians reveals activation of glycolysis during regeneration
Abstract Planarians are outstanding models for studying mechanisms of regeneration; however, there are few methods to measure changes in their metabolism. Examining metabolism in planarians is important because the regenerative process is dependent on numerous integrated metabolic pathways, which provide the energy required for tissue repair as well as the ability to synthesize the cellular building blocks needed to form new tissue. Therefore, we standardized an extracellular flux analysis method to measure mitochondrial and glycolytic activity in live planarians during normal growth as well as during regeneration. Small, uninjured planarians showed higher rates of oxygen consumption compared with large planarians, with no difference in glycolytic activity; however, glycolysis increased during planarian regeneration. Exposure of planarians to koningic acid, a specific inhibitor of glyceraldehyde‐3‐phosphate dehydrogenase, completely abolished extracellular acidification with little effect on oxygen consumption, which suggests that the majority of glucose catabolized in planarians is fated for aerobic glycolysis. These studies describe a useful method for measuring respiration and glycolysis in planarians and provide data implicating changes in glucose metabolism in the regenerative response.
DOI: 10.1016/j.yjmcc.2022.08.002
2022
Cited 13 times
Glutamine uptake and catabolism is required for myofibroblast formation and persistence
Fibrosis and extracellular matrix remodeling are mediated by resident cardiac fibroblasts (CFs). In response to injury, fibroblasts activate, differentiating into specialized synthetic and contractile myofibroblasts producing copious extracellular matrix proteins (e.g., collagens). Myofibroblast persistence in chronic diseases, such as HF, leads to progressive cardiac dysfunction and maladaptive remodeling. We recently reported that an increase in αKG (alpha-ketoglutarate) bioavailability, which contributes to enhanced αKG-dependent lysine demethylase activity and chromatin remodeling, is required for myofibroblast formation. Therefore, we aimed to determine the substrates and metabolic pathways contributing to αKG biosynthesis and their requirement for myofibroblast formation.Stable isotope metabolomics identified glutaminolysis as a key metabolic pathway required for αKG biosynthesis and myofibroblast formation, therefore we tested the effects of pharmacologic inhibition (CB-839) or genetic deletion of glutaminase (Gls1-/-) on myofibroblast formation in both murine and human cardiac fibroblasts. We employed immunofluorescence staining, functional gel contraction, western blotting, and bioenergetic assays to determine the myofibroblast phenotype.Carbon tracing indicated enhanced glutaminolysis mediating increased αKG abundance. Pharmacological and genetic inhibition of glutaminolysis prevented myofibroblast formation indicated by a reduction in αSMA+ cells, collagen gel contraction, collagen abundance, and the bioenergetic response. Inhibition of glutaminolysis also prevented TGFβ-mediated histone demethylation and supplementation with cell-permeable αKG rescued the myofibroblast phenotype. Importantly, inhibition of glutaminolysis was sufficient to prevent myofibroblast formation in CFs isolated from the human failing heart.These results define glutaminolysis as necessary for myofibroblast formation and persistence, providing substantial rationale to evaluate several new therapeutic targets to treat cardiac fibrosis.
DOI: 10.1038/s42003-022-03919-3
2022
Cited 13 times
Biomimetic cardiac tissue culture model (CTCM) to emulate cardiac physiology and pathophysiology ex vivo
There is need for a reliable in vitro system that can accurately replicate the cardiac physiological environment for drug testing. The limited availability of human heart tissue culture systems has led to inaccurate interpretations of cardiac-related drug effects. Here, we developed a cardiac tissue culture model (CTCM) that can electro-mechanically stimulate heart slices with physiological stretches in systole and diastole during the cardiac cycle. After 12 days in culture, this approach partially improved the viability of heart slices but did not completely maintain their structural integrity. Therefore, following small molecule screening, we found that the incorporation of 100 nM tri-iodothyronine (T3) and 1 μM dexamethasone (Dex) into our culture media preserved the microscopic structure of the slices for 12 days. When combined with T3/Dex treatment, the CTCM system maintained the transcriptional profile, viability, metabolic activity, and structural integrity for 12 days at the same levels as the fresh heart tissue. Furthermore, overstretching the cardiac tissue induced cardiac hypertrophic signaling in culture, which provides a proof of concept for the ability of the CTCM to emulate cardiac stretch-induced hypertrophic conditions. In conclusion, CTCM can emulate cardiac physiology and pathophysiology in culture for an extended time, thereby enabling reliable drug screening.
DOI: 10.1080/15216540701196944
2007
Cited 46 times
Role of glutathiolation in preservation, restoration and regulation of protein function
Abstract Glutathiolation has emerged as an important post‐translational modification that regulates protein function. Reduced glutathione remains bound to reactive cysteine side chains of several intracellular proteins even under basal conditions and the abundance of glutathiolated proteins increases upon oxidant challenge. Although protein glutathiolation was considered primarily to be a protective mechanism for preventing irreversible oxidation of protein thiols, recent evidence suggests that controlled glutathiolation reactions can also be used to modify protein structure and function. Several growth factors and cytokines promote protein glutathiolation and glutathiolated proteins have been shown to increase upon physiological stimulation of NO production. Given the high affinity of some nitrosylated proteins for glutathione, glutathiolation may also be a significant metabolic fate of nitrosylated proteins. Enzymatic pathways of protein de‐glutathiolation have also been described; indicating that both glutathiolation and de‐glutathiolation may be tightly regulated processes. In this review, we discuss the mechanisms of protein glutathiolation and how physiologic glutathiolation of specific proteins could regulate glucose metabolism, calcium homeostasis and changes in cell shape and contraction. We propose that glutathiolation represents a discrete sub‐state within complex thiol‐based redox circuits, relays and switches that regulate protein function basally and, upon oxidative stress, elicit adaptive responses or trigger cell death. IUBMB Life, 59: 21‐26, 2007
DOI: 10.1042/bj20081615
2008
Cited 46 times
Myocardial ischaemia inhibits mitochondrial metabolism of 4-hydroxy-<i>trans</i>-2-nonenal
Myocardial ischaemia is associated with the generation of lipid peroxidation products such as HNE (4-hydroxy-trans-2-nonenal); however, the processes that predispose the ischaemic heart to toxicity by HNE and related species are not well understood. In the present study, we examined HNE metabolism in isolated aerobic and ischaemic rat hearts. In aerobic hearts, the reagent [3H]HNE was glutathiolated, oxidized to [3H]4-hydroxynonenoic acid, and reduced to [3H]1,4-dihydroxynonene. In ischaemic hearts, [3H]4-hydroxynonenoic acid formation was inhibited and higher levels of [3H]1,4-dihydroxynonene and [3H]GS-HNE (glutathione conjugate of HNE) were generated. Metabolism of [3H]HNE to [3H]4-hydroxynonenoic acid was restored upon reperfusion. Reperfused hearts were more efficient at metabolizing HNE than non-ischaemic hearts. Ischaemia increased the myocardial levels of endogenous HNE and 1,4-dihydroxynonene, but not 4-hydroxynonenoic acid. Isolated cardiac mitochondria metabolized [3H]HNE primarily to [3H]4-hydroxynonenoic acid and minimally to [3H]1,4-dihydroxynonene and [3H]GS-HNE. Moreover, [3H]4-hydroxynonenoic acid was extruded from mitochondria, whereas other [3H]HNE metabolites were retained in the matrix. Mitochondria isolated from ischaemic hearts were found to contain 2-fold higher levels of protein-bound HNE than the cytosol, as well as increased [3H]GS-HNE and [3H]1,4-dihydroxynonene, but not [3H]4-hydroxynonenoic acid. Mitochondrial HNE oxidation was inhibited at an NAD+/NADH ratio of 0.4 (equivalent to the ischaemic heart) and restored at an NAD+/NADH ratio of 8.6 (equivalent to the reperfused heart). These results suggest that HNE metabolism is inhibited during myocardial ischaemia owing to NAD+ depletion. This decrease in mitochondrial metabolism of lipid peroxidation products and the inability of the mitochondria to extrude HNE metabolites could contribute to myocardial ischaemia/reperfusion injury.
DOI: 10.1016/s0076-6879(10)73009-3
2010
Cited 43 times
Measurement and Identification of S-Glutathiolated Proteins
Protein thiol modifications occur under both physiological and pathological conditions and can regulate protein function, redox signaling, and cell viability. The thiolation of proteins by glutathione (GSH) appears to be a particularly important mode of posttranslational modification that is increased under conditions of oxidative or nitrosative stress. Modification of proteins by glutathiolation has been shown to affect the structure and function of several susceptible proteins and protect them from subsequent oxidative injury. In many cases, the glutathiolated proteins are low in abundance, and dethiolation occurs readily. Therefore, sensitive, reliable, and reproducible methods are required for measuring both the total levels of protein glutathiolation and for identifying glutathiolated proteins under given conditions. These methods necessitate the preservation or the controlled removal of the GSH adducts during sample preparation for the accurate measurement of total S-glutathiolation and for the identification of protein-GSH adducts. In this chapter, we briefly review and provide protocols for chemical, mass spectrometric, immunological, and radioactive tagging techniques, for measuring protein S-glutathiolation in cells and tissues.
DOI: 10.1371/journal.pone.0175591
2017
Cited 29 times
Identification of a plasma metabolomic signature of thrombotic myocardial infarction that is distinct from non-thrombotic myocardial infarction and stable coronary artery disease
Current non-invasive diagnostics for acute myocardial infarction (MI) identify myocardial necrosis rather than the primary cause and therapeutic target-plaque disruption and resultant thrombosis. The aim of this study was to identify changes specific to plaque disruption and pathological thrombosis that are distinct from acute myocardial necrosis.We quantified 1,032 plasma metabolites by mass spectrometry in 11 thrombotic MI, 12 non-thrombotic MI, and 15 stable coronary artery disease (CAD) subjects at two acute phase (time of catheterization [T0], six hours [T6]) and one quiescent (>3 months follow-up) time points. A statistical classifier was constructed utilizing baseline (T0) abundances of a parsimonious set of 17 qualifying metabolites. Qualifying metabolites were those that demonstrated a significant change between the quiescent phase and the acute phase and that were distinct from any change seen in non-thrombotic MI or stable CAD subjects. Classifier performance as estimated by 10-fold cross-validation was suggestive of high sensitivity and specificity for differentiating thrombotic from non-thrombotic MI and stable CAD subjects at presentation. Nineteen metabolites demonstrated an intra-subject change from time of acute thrombotic MI presentation to the quiescent state that was distinct from any change measured in both the non-thrombotic MI and stable CAD subjects undergoing cardiac catheterization over the same time course (false discovery rate <5%).We have identified a candidate metabolic signature that differentiates acute thrombotic MI from quiescent state after MI, from acute non-thrombotic MI, and from stable CAD. Further validation of these metabolites is warranted given their potential as diagnostic biomarkers and novel therapeutic targets for the prevention or treatment of acute MI.
DOI: 10.1016/j.taap.2020.115213
2020
Cited 23 times
Heart slice culture system reliably demonstrates clinical drug-related cardiotoxicity
The limited availability of human heart tissue and its complex cell composition are major limiting factors for the reliable testing of drug efficacy and toxicity. Recently, we developed functional human and pig heart slice biomimetic culture systems that preserve the viability and functionality of 300 μm heart slices for up to 6 days. Here, we tested the reliability of this culture system for testing the cardiotoxicity of anti-cancer drugs. We tested three anti-cancer drugs (doxorubicin, trastuzumab, and sunitinib) with known different mechanisms of cardiotoxicity at three concentrations and assessed the effect of these drugs on heart slice viability, structure, function and gene expression. Slices incubated with any of these drugs for 48 h showed diminished in viability as well as loss of cardiomyocyte structure and function. Mechanistically, RNA sequencing of doxorubicin-treated tissues demonstrated a significant downregulation of cardiac genes and upregulation of oxidative stress responses. Trastuzumab treatment downregulated cardiac muscle contraction-related genes consistent with its clinically known effect on cardiomyocytes. Interestingly, sunitinib treatment resulted in significant downregulation of angiogenesis-related genes, in line with its mechanism of action. Similar to hiPS-derived-cardiomyocytes, heart slices recapitulated the expected toxicity of doxorubicin and trastuzumab, however, slices were superior in detecting sunitinib cardiotoxicity and mechanism in the clinically relevant concentration range of 0.1-1 μM. These results indicate that heart slice culture models have the potential to become a reliable platform for testing and elucidating mechanisms of drug cardiotoxicity.
DOI: 10.1161/circresaha.109.209791
2009
Cited 36 times
Beyond Reactive Oxygen Species
HomeCirculation ResearchVol. 105, No. 11Beyond Reactive Oxygen Species Free AccessEditorialPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessEditorialPDF/EPUBBeyond Reactive Oxygen SpeciesAldehydes as Arbitrators of Alarm and Adaptation Bradford G. Hill and Aruni Bhatnagar Bradford G. HillBradford G. Hill From the Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, Ky. Search for more papers by this author and Aruni BhatnagarAruni Bhatnagar From the Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, Ky. Search for more papers by this author Originally published20 Nov 2009https://doi.org/10.1161/CIRCRESAHA.109.209791Circulation Research. 2009;105:1044–1046Reactive oxygen species (ROS) are generated either during oxidative metabolism or in defense against pathogens. Their production is increased further by tissue injury or disease. Extensive evidence accumulated during the last 30 years provides compelling evidence that ROS-induced damage is a significant cause of cardiovascular injury and dysfunction. A variety of enzymatic and nonenzymatic antioxidants have evolved to protect against the constant onslaught of ROS, and these defenses respond deftly to changes in ROS generation or to the generation of secondary oxidation products. In this issue of Circulation Research, Endo et al1 report that increased mitochondrial accumulation of aldehydes derived from lipid oxidation protects against myocardial ischemia/reperfusion injury. These findings put a new twist on our view of the oxidant–antioxidant balance in the heart and underscore the importance of hormesis in which mild exposure to a stressor elicits an adaptive response that increases resistance to subsequent stress. Understanding how endogenous defense mechanisms are enhanced by stress response signaling could lead to the development of new strategies for combating oxidative stress.Most biological ROS are highly reactive and, therefore, short-lived. The ones with the greatest reactivity, such as the hydroxyl and alkoxyl radicals, have the shortest life span. As a result, the damage they induce is likely to be restricted to their site of origin and, therefore, of limited significance. Hence, it has long been suspected that ROS generate secondary products that spread injury and amplify damage. Which molecules can amplify and propagate ROS-initiated injury? Although many possibilities exist, one likely class of molecules may be aldehydes generated by the oxidation of unsaturated lipids. With the exception of antioxidants such as vitamin E or glutathione, unsaturated lipids are the most likely targets of ROS. When oxidized, these lipids generate a plethora of bioactive molecules, of which aldehydes are among the most reactive and abundant products. Being engendered by the violent fragmentation of their lipid parents, these aldehydes possess high reactivity. However, they are more stable than ROS, so they can diffuse to sites distant from their site of injury, thereby propagating oxidative injury. Moreover, these aldehydes possess a rich variety of structural features or they acquire additional ones by conjugating with receptive nucleophiles, which allows them to be recognized by cell constituents as signaling molecules. Aldehydes generated from oxidized lipids such as malondialdehyde, 4-hydroxy-trans-2-nonenal (HNE), and 1-palmitoyl-2-oxovaleroyl phosphatidyl choline (POVPC) have been detected in almost all tissues that have experienced oxidative injury. They are found in ischemic, hypertrophic, and failing hearts; in atherosclerotic lesions and restenotic vessels; in apoptotic cells; and in damaged mitochondria.2 Nevertheless, it is unclear whether they are simply markers of oxidative stress (footprints of a disruptive presence long gone) or whether they are active instruments of injury and sounders of alarm signaling.How does one study the active role of aldehydes in adaptation or injury? One approach is to examine aldehyde metabolism. Like ROS, aldehydes are metabolized and detoxified by several nonenzymatic and enzymatic processes (see the Figure). Previous work has shown that in most cells aldehydes are either reduced (to alcohols), oxidized (to acids), or conjugated with cellular nucleophiles such as glutathione, carnosine (β-alanine-l-histidine), or proteins. Several enzymes involved in metabolizing these aldehydes have been identified.2 In cardiovascular tissue, aldose reductase (AR) catalyzes the reduction of both HNE and POVPC. Oxidation of aldehydes is catalyzed by aldehyde dehydrogenases (ALDHs) and glutathione conjugation is facilitated by glutathione S-transferases. By converting aldehydes to less reactive products, these enzymes prevent the direct toxicity of aldehydes; however, metabolic conversion could also enhance stability of aldehydes and thereby augment their ability to stimulate cell signaling. Hence, investigations into the role of aldehyde-metabolizing enzymes in modulating outcomes of oxidative injury could provide one avenue for understanding the role of aldehydes as mediators of oxidative stress–related signaling. Download figureDownload PowerPointFigure. Pathways of aldehyde-induced stress responses in the cardiovascular system. The peroxidation of membrane lipids by ROS leads to the formation of aldehydes (RCHO). Aldehydes are detoxified by multiple enzymes in the cell; they can be reduced by aldo-keto reductases (AR) to their corresponding alcohols (RCH2OH), oxidized to carboxylates via ALDHs, or conjugated to glutathione (GSH) or l-carnosine to form glutathione conjugates (GS-RCHO) and l-carnosine conjugates (His-RCHO), respectively. The glutathione conjugation reactions are known to be catalyzed by GST. Reduction of the conjugate by AR has been shown to promote inflammation. Aldehydes can deplete glutathione, modify proteins, and promote mitochondrial dysfunction and nutrient stress, which may cause ER stress. ER stress can promote the activation of adaptive arms of the unfolded protein response, leading to induction of proteins, ie, ATF4 and -6, associated with metabolic remodeling and a cardioprotected phenotype. ER stress can also lead to inflammatory responses, which, alone or in combination with mitochondrial dysfunction, could increase atherosclerotic lesion formation or promote myocardial ischemia/reperfusion injury.In keeping with the view that ROS-mediated injury could be attributed in part to lipid-derived aldehydes, it has been shown that modulation of aldehyde metabolism profoundly affects cardiovascular injury. For example, inhibition of the aldehyde-metabolizing enzyme AR abolishes the late phase of ischemic preconditioning3 and exacerbates atherogenesis.4 Moreover, treatment with small molecule activators of ALDH2 to stimulate the metabolism of cytotoxic aldehydes reduces infarct size by 60%,5 indicating that much of the damage inflicted by ischemia/reperfusion could be attributed to aldehydes generated in the ischemic heart. In contrast, Endo et al1 report that transgenic expression of an Aldh2 gene containing a single nucleotide polymorphism (Aldh2*2), with impaired ALDH activity, diminishes ischemic injury. They reconcile these results by postulating that mitochondrial aldehyde accumulation induces a hormetic response that leads to a cardioprotected phenotype.The Aldh2*2 allele is present in a large proportion of people of Asian descent and results in a frank inability to tolerate alcohol. In addition to alcohol flushing syndrome, this mutation results in an increase in the levels of serum peroxide and an increase in the risk of late-onset Alzheimer’s disease. Few other physiological effects of this mutation have been reported. Intriguingly, transgenic expression Aldh2*2 in the skeletal muscle and hearts of mice resulted in stunted growth, reduced muscle mass and fat content, osteopenia, and kyphosis. Expression of Aldh2*2 decreased the ability of the heart to metabolize HNE, with extensive metabolic remodeling. Surprisingly, no significant changes in the expression of most of the major antioxidant enzymes were observed, but there was a shift in glucose metabolism from glycolysis to the pentose phosphate pathway. These changes seem to be related to the activation of the eukaryotic initiation factor (eIF)2α–activating transcription factor (ATF)4 pathway, because heterozygous knockout of ATF4 blunted the increase in glutathione in the Aldh2*2 hearts and weakened the ischemia-resistant phenotype.The eIF2α-ATF4 pathway is a regulator of amino acid metabolism and is activated as a component of the unfolded protein response, which is triggered by the accumulation of unfolded proteins in the endoplasmic reticulum (ER).6 Because transgenic expression of Aldh2*2 was associated with an increase in the accumulation of protein-HNE adducts, it is likely that that HNE-adducted proteins could trigger unfolded protein response pathways. Further studies are required to elucidate the role of ER stress in aldehyde-induced hormesis; however, the findings of Endo et al1 reveal novel pathways of crosstalk between mitochondrial stress, antioxidant defense, and intermediary metabolism. In our present understanding of ischemic preconditioning, brief bouts of ischemia increase cytosolic cardioprotective proteins, which then protect the mitochondria during lethal episodes of ischemia. Here, stress originating primarily from mitochondria established cardioprotection by altering cytosolic metabolism, indicating a new mechanism for sensing mitochondrial stress related to the activation of ATF4.Although the efficacy of sublethal stress in establishing protection has been demonstrated in a variety of experimental paradigms (eg, ischemic preconditioning, calorie restriction), the true value of such Spartan principles is only now beginning to emerge. For instance, it has been reported recently that decreasing glucose metabolism extends the life span of Caenorhabditis elegans because it increases mitochondrial ROS production.7 Significantly, treatment with antioxidants and vitamins prevented the extension of life span, arguing against many commonly held beliefs in the utility of antioxidant supplements and providing an additional reason why clinical trials with vitamin supplements have been so disappointingly negative.The observation that ischemic resistance of Aldh2*2 transgenic hearts was related to metabolic remodeling also provides new support to the concept that antioxidant defense could be strengthened, not only by elevating the levels of antioxidants (vitamins C and E) or antioxidant enzymes (superoxide dismutase or catalase) but also by increasing metabolic flux through pathways that generate reducing equivalents (NADPH and glutathione). That this protection was achieved by rechanneling glucose from glycolysis to the shunt and by changing amino acid metabolism points to the presence of ancient stress-activated circuits linking nutrition and energy metabolism to survival and self-defense.It has long been suspected that glucose metabolism has a primary role in antioxidant defense. However, glucose could have opposing effects: it could increase ROS production (by increasing metabolism) or prevent oxidative injury (by providing reducing equivalents). Endo et al1 were able to uncover an unambiguous role of glucose in cardioprotection because they were looking at the effects of secondary products rather than ROS themselves. Direct exposure to ROS alters many things simultaneously and as a result it is difficult to distinguish cause from effect or primary from secondary changes. Surprisingly, they found that instead of an increase in the levels of the usual suspects (catalase, glutathione peroxidases, reductases, superoxide dismutases), there was an upregulation of glycine and serine metabolism and an ATF4-mediated increase in glutathione synthesis. These observations provide fresh support to the idea that to protect cells from oxidative stress-related injury it may be necessary to increase the metabolism of lipid-derived aldehydes rather than to simply increase ROS quenching.That increased generation of aldehydes could trigger extensive and complex remodeling in the heart attests to the high biological activity of lipid-derived aldehydes and their unique ability to trigger stress signaling. Nevertheless, these conclusions are based on strategies that alter aldehyde metabolism. Elucidation of the in vivo role of aldehydes by altering their metabolism is, however, not straightforward. For example, AR which metabolizes aldehydes also reduces glucose to sorbitol. Hence, some of the effects observed with changes in AR levels may relate to changes in glucose metabolism. Similarly, ALDH2 not only metabolizes HNE but several other aldehydes as well, and it is significant that in the study by Endo et al1 the Aldh2*2 transgenic hearts were smaller in size suggesting that the enzyme may be necessary for growth-regulating metabolism or that changes in the metabolism of aldehydes in the skeletal muscle (where the Aldh2*2 transgene was also expressed) could indirectly affect myocardial sensitivity to ischemia. Moreover, complete deletion of the Aldh2 gene did not affect aldehyde metabolism, indicating that this enzyme does not metabolize aldehydes in vivo. Aldehyde metabolism was, however, affected in Aldh2*2 hearts, which the authors speculate may be attributable to its ability to bind to other ALDHs. If ALDH2 is not the main enzyme catalyzing aldehyde oxidation, then why does the activation of this enzyme by small molecule activators prevent ischemic injury5? Clearly, like all studies of merit, the study by Endo et al raises more questions than it answers. Nonetheless, the present findings are intriguing and strengthen the rationale for studying whether the presence of the Aldh2*2 allele in humans is associated with an increase in resistance to ischemic heart disease or other cardiovascular diseases associated with an increase in ROS production. Table 1. Non-standard Abbreviations and AcronymsALDHaldehyde dehydrogenaseARaldose reductaseATFactivating transcription factoreIFeukaryotic initiation factorERendoplasmic reticulumHNE4-hydroxy-trans-2-nonenalPOVPC1-palmitoyl-2-oxovaleroyl phosphatidyl cholineROSreactive oxygen speciesThe opinions expressed in this editorial are not necessarily those of the editors or of the American Heart Association.Sources of FundingSupported by National Heart, Lung, and Blood Institute (NIH) grants HL55477, HL59378, and HL078825 and National Center for Research Researches (NIH) grant RR24489.DisclosuresNone.FootnotesCorrespondence to Aruni Bhatnagar, Diabetes and Obesity Center, University of Louisville, Delia Baxter Building, Rm 421, 580 South Preston St, Louisville, KY 40202. E-mail [email protected] References 1 Endo J, Sano M, Katayama T, Hishiki T, Shinmura K, Morizane S, Matsuhashi T, Katsumata Y, Zhang Y, Ito H, Nagahata Y, Marchitti S, Nishimaki K, Wolf AM, Nakanishi H, Hattori F, Vasiliou V, Adachi T, Ohsawa I, Taguchi R, Hirabayashi Y, Ohta S, Suematsu M, Ogawa S, Fukuda K. Metabolic remodeling induced by mitochondrial aldehyde stress stimulates tolerance to oxidative stress in the heart. Circ Res. 2009; 105: 1118–1127.LinkGoogle Scholar2 Conklin D, Prough R, Bhatanagar A. Aldehyde metabolism in the cardiovascular system. Mol Biosyst. 2007; 3: 136–150.CrossrefMedlineGoogle Scholar3 Shinmura K, Bolli R, Liu SQ, Tang XL, Kodani E, Xuan YT, Srivastava S, Bhatnagar A. Aldose reductase is an obligatory mediator of the late phase of ischemic preconditioning. Circ Res. 2002; 91: 240–246.LinkGoogle Scholar4 Srivastava S, Vladykovskaya E, Barski OA, Spite M, Kaiserova K, Petrash JM, Chung SS, Hunt G, Dawn B, Bhatnagar A. Aldose reductase protects against early atherosclerotic lesion formation in apolipoprotein E-null mice. Circ Res. 2009; 105: 793–802.LinkGoogle Scholar5 Chen CH, Budas GR, Churchill EN, Disatnik MH, Hurley TD, Mochly-Rosen D. Activation of aldehyde dehydrogenase-2 reduces ischemic damage to the heart. Science. 2008; 321: 1493–1495.CrossrefMedlineGoogle Scholar6 Xu C, Bailly-Maitre B, Reed JC. Endoplasmic reticulum stress: cell life and death decisions. J Clin Invest. 2005; 115: 2656–2664.CrossrefMedlineGoogle Scholar7 Schulz TJ, Zarse K, Voigt A, Urban N, Birringer M, Ristow M. Glucose restriction extends Caenorhabditis elegans life span by inducing mitochondrial respiration and increasing oxidative stress. Cell Metab. 2007; 6: 280–293.CrossrefMedlineGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetailsCited By Bai Y, Wang M, Yang Y, Liu X, Chen Q and Guo Z (2021) Inhibition of the miR‐193b‐3p protects against oxidized low‐density lipoprotein‐induced HUVECs injury by upregulating ALDH2, Cell Biology International, 10.1002/cbin.11720, 46:2, (192-202), Online publication date: 1-Feb-2022. Biagini D, Lomonaco T, Ghimenti S, Fusi J, Cerri E, De Angelis F, Bellagambi F, Oger C, Galano J, Bramanti E, Franzoni F, Fuoco R and Di Francesco F (2020) Saliva as a non-invasive tool for monitoring oxidative stress in swimmers athletes performing a VO2max cycle ergometer test, Talanta, 10.1016/j.talanta.2020.120979, 216, (120979), Online publication date: 1-Aug-2020. Erban T, Sopko B, Kadlikova K, Talacko P and Harant K (2019) Varroa destructor parasitism has a greater effect on proteome changes than the deformed wing virus and activates TGF-β signaling pathways, Scientific Reports, 10.1038/s41598-019-45764-1, 9:1, Online publication date: 1-Dec-2019. Hua T, Yang M, Zhou Y, Chen L, Wu H and Liu R (2019) Alda-1 Prevents Pulmonary Epithelial Barrier Dysfunction following Severe Hemorrhagic Shock through Clearance of Reactive Aldehydes, BioMed Research International, 10.1155/2019/2476252, 2019, (1-9), Online publication date: 4-Aug-2019. Dator R, Solivio M, Villalta P and Balbo S (2019) Bioanalytical and Mass Spectrometric Methods for Aldehyde Profiling in Biological Fluids, Toxics, 10.3390/toxics7020032, 7:2, (32) Badran M, Abuyassin B, Golbidi S, Ayas N and Laher I (2019) Alpha Lipoic Acid Improves Endothelial Function and Oxidative Stress in Mice Exposed to Chronic Intermittent Hypoxia, Oxidative Medicine and Cellular Longevity, 10.1155/2019/4093018, 2019, (1-13), Online publication date: 9-Apr-2019. Conklin D, Guo Y, Nystoriak M, Jagatheesan G, Obal D, Kilfoil P, Hoetker J, Guo L, Bolli R and Bhatnagar A (2019) TRPA1 channel contributes to myocardial ischemia-reperfusion injury, American Journal of Physiology-Heart and Circulatory Physiology, 10.1152/ajpheart.00106.2018, 316:4, (H889-H899), Online publication date: 1-Apr-2019. YASUE H, MIZUNO Y and HARADA E (2019) Coronary artery spasm — Clinical features, pathogenesis and treatment —, Proceedings of the Japan Academy, Series B, 10.2183/pjab.95.005, 95:2, (53-66), Online publication date: 8-Feb-2019. Yasue H, Mizuno Y and Harada E (2019) Association of East Asian Variant Aldehyde Dehydrogenase 2 Genotype (ALDH2*2*) with Coronary Spasm and Acute Myocardial Infarction Aldehyde Dehydrogenases, 10.1007/978-981-13-6260-6_7, (121-134), . Ding J, Yang Z, Ma H and Zhang H (2019) Mitochondrial Aldehyde Dehydrogenase in Myocardial Ischemic and Ischemia-Reperfusion Injury Aldehyde Dehydrogenases, 10.1007/978-981-13-6260-6_6, (107-120), . Tango T, Ichwan S, Wanandi S and Hardiany N (2018) Relative expression of aldehyde dehydrogenase 1 family member A1 in different malignancies of human glioma cells, Journal of Physics: Conference Series, 10.1088/1742-6596/1073/3/032059, 1073, (032059), Online publication date: 1-Aug-2018. Dassanayaka S, Zheng Y, Gibb A, Cummins T, McNally L, Brittian K, Jagatheesan G, Audam T, Long B, Brainard R, Jones S and Hill B (2018) Cardiac-specific overexpression of aldehyde dehydrogenase 2 exacerbates cardiac remodeling in response to pressure overload, Redox Biology, 10.1016/j.redox.2018.05.016, 17, (440-449), Online publication date: 1-Jul-2018. Yang M, Wang Y, Han B, Yang B, Qiang Y, Zhang Y, Wang Z, Huang X, Liu J, Chen Y, Ren J, Cao F and Xu Y (2017) Activation of aldehyde dehydrogenase 2 slows down the progression of atherosclerosis via attenuation of ER stress and apoptosis in smooth muscle cells, Acta Pharmacologica Sinica, 10.1038/aps.2017.81, 39:1, (48-58), Online publication date: 1-Jan-2018. Pérez-Alea M, McGrail K, Sánchez-Redondo S, Ferrer B, Fournet G, Cortés J, Muñoz E, Hernandez-Losa J, Tenbaum S, Martin G, Costello R, Ceylan I, Garcia-Patos V and Recio J (2017) ALDH1A3 is epigenetically regulated during melanocyte transformation and is a target for melanoma treatment, Oncogene, 10.1038/onc.2017.160, 36:41, (5695-5708), Online publication date: 12-Oct-2017. Gasparetto M and Smith C (2017) ALDHs in normal and malignant hematopoietic cells: Potential new avenues for treatment of AML and other blood cancers, Chemico-Biological Interactions, 10.1016/j.cbi.2017.06.020, 276, (46-51), Online publication date: 1-Oct-2017. Mizuno Y, Hokimoto S, Harada E, Kinoshita K, Yoshimura M and Yasue H (2017) Variant Aldehyde Dehydrogenase 2 (ALDH2*2) in East Asians Interactively Exacerbates Tobacco Smoking Risk for Coronary Spasm ― Possible Role of Reactive Aldehydes ―, Circulation Journal, 10.1253/circj.CJ-16-0969, 81:1, (96-102), . Li M, Zhang P, Wei H, Li M, Zou W, Li X, Gu H and Tang X (2016) Hydrogen Sulfide Ameliorates Homocysteine-Induced Cognitive Dysfunction by Inhibition of Reactive Aldehydes Involving Upregulation of ALDH2, International Journal of Neuropsychopharmacology, 10.1093/ijnp/pyw103, (pyw103) Ding J, Zhang Q, Luo Q, Ying Y, Liu Y, Li Y, Wei W, Yan F and Zhang H (2016) Alda-1 Attenuates Lung Ischemia-Reperfusion Injury by Reducing 4-Hydroxy-2-Nonenal in Alveolar Epithelial Cells, Critical Care Medicine, 10.1097/CCM.0000000000001563, 44:7, (e544-e552), Online publication date: 1-Jul-2016. Mizuno Y, Hokimoto S, Harada E, Kinoshita K, Nakagawa K, Yoshimura M, Ogawa H and Yasue H (2016) Variant Aldehyde Dehydrogenase 2 (2*2) Is a Risk Factor for Coronary Spasm and ST‐Segment Elevation Myocardial Infarction, Journal of the American Heart Association, 5:5, Online publication date: 6-May-2016.Conklin D, Guo Y, Jagatheesan G, Kilfoil P, Haberzettl P, Hill B, Baba S, Guo L, Wetzelberger K, Obal D, Rokosh D, Prough R, Prabhu S, Velayutham M, Zweier J, Hoetker J, Riggs D, Srivastava S, Bolli R and Bhatnagar A (2015) Genetic Deficiency of Glutathione S-Transferase P Increases Myocardial Sensitivity to Ischemia–Reperfusion Injury, Circulation Research, 117:5, (437-449), Online publication date: 14-Aug-2015. Guo J, Liu A, Zang P, Dong W, Ying L, Wang W, Xu P, Song X, Cai J, Zhang S, Duan J, Mehta J and Su D (2013) ALDH2 protects against stroke by clearing 4-HNE, Cell Research, 10.1038/cr.2013.69, 23:7, (915-930), Online publication date: 1-Jul-2013. Gong D, Zhang H and Hu S (2013) Mitochondrial aldehyde dehydrogenase 2 activation and cardioprotection, Journal of Molecular and Cellular Cardiology, 10.1016/j.yjmcc.2012.03.017, 55, (58-63), Online publication date: 1-Feb-2013. Gong D, Zhang Y, Zhang H, Gu H, Jiang Q and Hu S (2012) Aldehyde Dehydrogenase-2 Activation during Cardioplegic Arrest Enhances the Cardioprotection against Myocardial Ischemia–Reperfusion Injury, Cardiovascular Toxicology, 10.1007/s12012-012-9179-6, 12:4, (350-358), Online publication date: 1-Dec-2012. Luo F, Thiele B, Janzik I, Zeng B, Schurr U and Matsubara S (2012) De-submergence responses of antioxidative defense systems in two wetland plants having escape and quiescence strategies, Journal of Plant Physiology, 10.1016/j.jplph.2012.06.015, 169:17, (1680-1689), Online publication date: 1-Nov-2012. Zhang H, Gong D, Zhang Y, Li S and Hu S (2012) Effect of mitochondrial aldehyde dehydrogenase-2 genotype on cardioprotection in patients with congenital heart disease, European Heart Journal, 10.1093/eurheartj/ehs061, 33:13, (1606-1614), Online publication date: 1-Jul-2012., Online publication date: 1-Jul-2012. Lin Z, Li H, Luo H, Zhang Y and Luo W (2011) Benzylamine and methylamine, substrates of semicarbazide-sensitive amine oxidase, attenuate inflammatory response induced by lipopolysaccharide, International Immunopharmacology, 10.1016/j.intimp.2011.03.002, 11:8, (1079-1089), Online publication date: 1-Aug-2011. Zhang Y, Sano M, Shinmura K, Tamaki K, Katsumata Y, Matsuhashi T, Morizane S, Ito H, Hishiki T, Endo J, Zhou H, Yuasa S, Kaneda R, Suematsu M and Fukuda K (2010) 4-Hydroxy-2-nonenal protects against cardiac ischemia–reperfusion injury via the Nrf2-dependent pathway, Journal of Molecular and Cellular Cardiology, 10.1016/j.yjmcc.2010.05.011, 49:4, (576-586), Online publication date: 1-Oct-2010. November 20, 2009Vol 105, Issue 11 Advertisement Article InformationMetrics https://doi.org/10.1161/CIRCRESAHA.109.209791PMID: 19926880 Originally publishedNovember 20, 2009 PDF download Advertisement
DOI: 10.1016/j.cbi.2008.10.055
2009
Cited 35 times
Aldose reductase decreases endoplasmic reticulum stress in ischemic hearts
Aldose reductase (AR) is a multi-functional AKR (AKR1B1) that catalyzes the reduction of a wide range of endogenous and xenobiotic aldehydes and their glutathione conjugates with high efficiency. Previous studies from our laboratory show that AR protects against myocardial ischemia-reperfusion injury, however, the mechanisms by which it confers cardioprotection remain unknown. Because AR metabolizes aldehydes generated from lipid peroxidation, we tested the hypothesis that it protects against ischemic injury by preventing ER stress induced by excessive accumulation of aldehyde-modified proteins in the ischemic heart. In cell culture experiments, exposure to model lipid peroxidation aldehydes-4-hydroxy-trans-2-nonenal (HNE), 1-palmitoyl-2-oxovaleroyl phosphatidylcholine (POVPC) or acrolein led to an increase in the phosphorylation of ER stress markers PERK and eIF2-alpha and an increase in ATF3. The reduced metabolite of POVPC 1-palmitoyl-2-hydroxyvaleroyl phosphatidylcholine (PHVPC) was unable to stimulate JNK phosphorylation. No increase in phospho-eIF2-alpha, ATF3 or phospho-PERK was observed in cells treated with the reduced HNE metabolite 1,4-dihydroxynonenol (DHN). Lysates prepared from isolated perfused mouse hearts subjected to 15 min of global ischemia followed by 30 min of reperfusion ex vivo showed greater phosphorylation of PERK and eIF2-alpha than hearts subjected to aerobic perfusion alone. Ischemia-induced increases in phospho-PERK and phospho-eIF2-alpha were diminished in the hearts of cardiomyocyte-specific transgenic mice overexpressing the AR transgene. These observations support the notion that by removing aldehydic products of lipid peroxidation, AR decreases ischemia-reperfusion injury by diminishing ER stress.
DOI: 10.1016/j.redox.2013.07.001
2013
Cited 30 times
Quercetin prevents left ventricular hypertrophy in the Apo E knockout mouse
Hypercholesterolemia is a risk factor for the development of hypertrophic cardiomyopathy. Nevertheless, there are few studies aimed at determining the effects of dietary compounds on early or mild cardiac hypertrophy associated with dyslipidemia. Here we describe left ventricular (LV) hypertrophy in 12 week-old Apo E(-/-) hypercholesterolemic mice. The LV end diastolic posterior wall thickness and overall LV mass were significantly increased in Apo E(-/-) mice compared with wild type (WT) controls. Fractional shortening, LV end diastolic diameter, and hemodynamic parameters were unchanged from WT mice. Oral low dose quercetin (QCN; 0.1 µmol QCN/kg body weight for 6 weeks) significantly reduced total cholesterol and very low density lipoprotein in the plasma of Apo E(-/-) mice. QCN treatment also significantly decreased LV posterior wall thickness and LV mass in Apo E(-/-) mice. Myocardial geometry and function were unaffected in WT mice by QCN treatment. These data suggest that dietary polyphenolic compounds such as QCN may be effective modulators of plasma cholesterol and could prevent maladaptive myocardial remodeling.
DOI: 10.1186/2044-5040-3-18
2013
Cited 30 times
TWEAK promotes exercise intolerance by decreasing skeletal muscle oxidative phosphorylation capacity
Proinflammatory cytokine tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) and its receptor Fn14 are the major regulators of skeletal muscle mass in many catabolic conditions. However, their role in muscle metabolism remains largely unknown. In the present study, we investigated the role of TWEAK on exercise capacity and skeletal muscle mitochondrial content and oxidative metabolism.We employed wild-type and TWEAK-knockout (KO) mice and primary myotube cultures and performed biochemical, bioenergetics, and morphometric assays to evaluate the effects of TWEAK on exercise tolerance and muscle mitochondrial function and angiogenesis.TWEAK-KO mice showed improved exercise tolerance compared to wild-type mice. Electron microscopy analysis showed that the abundance of subsarcolemmal and intermyofibrillar mitochondria is significantly increased in skeletal muscle of TWEAK-KO mice compared to wild-type mice. Furthermore, age-related loss in skeletal muscle oxidative capacity was rescued in TWEAK-KO mice. Expression of a key transcriptional regulator peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) and several other molecules involved in oxidative metabolism were significantly higher in skeletal muscle of TWEAK-KO mice. Moreover, treatment of primary myotubes with soluble TWEAK inhibited the expression of PGC-1α and mitochondrial genes and decreased mitochondrial respiratory capacity. Deletion of TWEAK also improved angiogenesis and transcript levels of vascular endothelial growth factor in skeletal muscle of mice.These results demonstrate that TWEAK decreases mitochondrial content and oxidative phosphorylation and inhibits angiogenesis in skeletal muscle. Neutralization of TWEAK is a potential approach for improving exercise capacity and oxidative metabolism in skeletal muscle.
DOI: 10.1139/o62-115
1962
Cited 24 times
THE UNCOUPLING OF OXIDATIVE PHOSPHORYLATION BY IONIZING RADIATION
Whole body irradiation of rabbits or rats with X-rays or Co 60 γ-rays causes uncoupling of oxidative phosphorylation in thymus mitochondria, which is not prevented by the prior administration of AET. Whole body irradiation was not found to affect oxidative phosphorylation in liver or mouse ascites cell mitochondria. The radiation lesion can be repaired in vitro by the addition of cytochrome c, bovine serum albumin, or vitamin K 1 to mitochondria. Vitamin E and coenzyme Q10 were without effect. Both phosphorylating steps in the electron transport chain associated with succinate oxidation are affected by irradiation. The diphosphopyridine nucleotide dependent steps in the oxidation of α-ketoglutarate by thymus mitochondria are damaged by in vivo irradiation. Diphosphopyridine nucleotide levels of thymus and spleen but not liver or ascites cells are reduced by in vivo irradiation. No effect of in vitro irradiation on oxidative phosphorylation could be found for thymocyte cell suspensions, isolated thymus or liver mitochondria, or ascites or HeLa cell suspensions. Respiration of ascites or thymocyte cells was unaffected by in vitro irradiation.
DOI: 10.3389/fphys.2016.00636
2016
Cited 23 times
FVB/NJ Mice Are a Useful Model for Examining Cardiac Adaptations to Treadmill Exercise
Mice are commonly used to examine the mechanisms by which exercise improves cardiometabolic health; however, exercise compliance and adaptations are often strain-dependent or are variable due to inconsistency in exercise training protocols. In this study, we examined nocturnal/diurnal behavior, treadmill exercise compliance, and systemic as well as cardiac-specific exercise adaptations in two commonly used mouse strains, C57BL/6J, and FVB/NJ mice. Metabolic cage analysis indicated a strong nocturnal nature of C57BL/6J mice, whereas FVB/NJ mice showed no circadian element to activity, food or water intake, VO2, or VCO2. Initial exercise capacity tests revealed that, compared with C57BL/6J mice, FVB/NJ mice are capable of achieving nearly 2-fold higher workloads prior to exhaustion. FVB/NJ mice tested during the day were capable of achieving significantly more work compared with their night-tested counterparts. Following 4 weeks of training, FVB/NJ mice showed significant increases in exercise capacity as well as physiologic cardiac growth characterized by enlarged myocytes and higher mitochondrial DNA content. C57BL/6J mice showed no increases in exercise capacity or cardiac growth regardless of whether they exercised during the day or the night. This lack of adaptation in C57BL/6J mice was attributable, at least in part, to their progressive loss of compliance to the treadmill training protocol. We conclude that the FVB/NJ strain is a useful and robust mouse model for examining cardiac adaptations to treadmill exercise and that treadmill training during daytime hours does not negatively affect exercise compliance or capacity.
DOI: 10.1152/ajpheart.00804.2020
2021
Cited 16 times
Endothelial progenitor cells as critical mediators of environmental air pollution-induced cardiovascular toxicity
Environmental air pollution exposure is a leading cause of death worldwide, and with increasing industrialization and urbanization, its disease burden is expected to rise even further. The majority of air pollution exposure-associated deaths are linked to cardiovascular disease (CVD). Although ample research demonstrates a strong correlation between air pollution exposure and CVD risk, the mechanisms by which inhalation of polluted air affects cardiovascular health are not completely understood. Inhalation of environmental air pollution has been associated with endothelial dysfunction, which suggests that air pollution exposure impacts CVD health by inducing endothelial injury. Interestingly, recent studies demonstrate that air pollution exposure affects the number and function of endothelial progenitor cells (EPCs), subpopulations of bone marrow-derived proangiogenic cells that have been shown to play an essential role in maintaining cardiovascular health. In line with their beneficial function, chronically low levels of circulating EPCs and EPC dysfunction (e.g., in diabetic patients) have been associated with vascular dysfunction, poor cardiovascular health, and increases in the severity of cardiovascular outcomes. In contrast, treatments that improve EPC number and function (e.g., exercise) have been found to attenuate cardiovascular dysfunction. Considering the critical, nonredundant role of EPCs in maintaining vascular health, air pollution exposure-induced impairments in EPC number and function could lead to endothelial dysfunction, consequently increasing the risk for CVD. This review article covers novel aspects and new mechanistic insights of the adverse effects of air pollution exposure on cardiovascular health associated with changes in EPC number and function.
DOI: 10.1016/j.redox.2021.102094
2021
Cited 16 times
Cell cycle induction in human cardiomyocytes is dependent on biosynthetic pathway activation
The coordinated gene and metabolic programs that facilitate cardiomyocyte entry and progression in the cell cycle are poorly understood. The purpose of this study was to identify the metabolic changes that influence myocyte proliferation.In adult mouse cardiomyocytes and human induced pluripotent stem cell cardiomyocytes (hiPS-CMs), cell cycle initiation by ectopic expression of Cyclin B1, Cyclin D1, CDK1, and CDK4 (termed 4F) downregulated oxidative phosphorylation genes and upregulated genes that regulate ancillary biosynthetic pathways of glucose metabolism. Results from metabolic analyses and stable isotope tracing experiments indicate that 4F-mediated cell cycle induction in hiPS-CMs decreases glucose oxidation and oxidative phosphorylation and augments NAD+, glycogen, hexosamine, phospholipid, and serine biosynthetic pathway activity. Interventions that diminish NAD+ synthesis, serine synthesis, or protein O-GlcNAcylation decreased 4F-mediated cell cycle entry. In a gain of function approach, we overexpressed phosphoenolpyruvate carboxykinase 2 (PCK2), which can drive carbon from the Krebs cycle to the glycolytic intermediate pool, and found that PCK2 augments 4F-mediated cell cycle entry.These findings suggest that a metabolic shift from catabolic to anabolic activity is a critical step for cardiomyocyte cell cycle entry and is required to facilitate proliferation.
DOI: 10.1152/ajpheart.00105.2022
2022
Cited 9 times
Metabolic signatures of pregnancy-induced cardiac growth
Little is known of the underlying molecular and cellular mechanisms that contribute to pregnancy-induced cardiac growth. Several lines of evidence suggest that changes in cardiac metabolism may contribute. Here, we provide a comprehensive metabolic atlas of the metabolomic, proteomic, and transcriptomic changes occurring in the maternal heart. We show that pregnancy-induced cardiac growth is associated with changes in glycerophospholipid, nucleotide, and amino acid metabolism, with reductions in cardiac glucose catabolism. Collectively, these results suggest that substantial metabolic changes occur in the maternal heart during pregnancy and after pregnancy.
DOI: 10.1016/j.jshs.2022.06.001
2022
Cited 9 times
Influence of biological sex and exercise on murine cardiac metabolism
Although the structural and functional effects of exercise on the heart are well established, the metabolic changes that occur in the heart during and after exercise remain unclear. In this study, we used metabolomics to assess time-dependent changes in the murine cardiac metabolome following 1 session of treadmill exercise. After the exercise bout, we also recorded blood lactate, glucose, and ketone body levels and measured cardiac mitochondrial respiration. In both male and female mice, moderate- and high-intensity exercise acutely increased blood lactate levels. In both sexes, low- and moderate-intensity exercise augmented circulating 3-hydroxybutryrate levels immediately after the exercise bout; however, only in female mice did high-intensity exercise increase 3-hydroxybutyrate levels, with significant increases occurring 1 h after the exercise session. Untargeted metabolomics analyses of sedentary female and male hearts suggest considerable sex-dependent differences in basal cardiac metabolite levels, with female hearts characterized by higher levels of pantothenate, pyridoxamine, homoarginine, tryptophan, and several glycerophospholipid and sphingomyelin species and lower levels of numerous metabolites, including acetyl coenzyme A, glucuronate, gulonate, hydroxyproline, prolyl-hydroxyproline, carnosine, anserine, and carnitinylated and glycinated species, as compared with male hearts. Immediately after a bout of treadmill exercise, both male and female hearts had higher levels of corticosterone; however, female mice showed more extensive exercise-induced changes in the cardiac metabolome, characterized by significant, time-dependent changes in amino acids (e.g., serine, alanine, tyrosine, tryptophan, branched-chain amino acids) and the ketone body 3-hydroxybutyrate. Results from experiments using isolated cardiac mitochondria suggest that high-intensity treadmill exercise does not acutely affect respiration or mitochondrial coupling; however, female cardiac mitochondria demonstrate generally higher adenosine diphosphate sensitivity compared with male cardiac mitochondria. Collectively, these findings in mice reveal key sex-dependent differences in cardiac metabolism and suggest that the metabolic network in the female heart is more responsive to physiological stress caused by exercise.
DOI: 10.1016/j.redox.2023.102740
2023
Cited 3 times
The metabolic state of the heart regulates mitochondrial supercomplex abundance in mice
Mitochondrial supercomplexes are observed in mammalian tissues with high energy demand and may influence metabolism and redox signaling. Nevertheless, the mechanisms that regulate supercomplex abundance remain unclear. In this study, we examined the composition of supercomplexes derived from murine cardiac mitochondria and determined how their abundance changes with substrate provision or by genetically induced changes to the cardiac glucose-fatty acid cycle. Protein complexes from digitonin-solubilized cardiac mitochondria were resolved by blue-native polyacrylamide gel electrophoresis and were identified by mass spectrometry and immunoblotting to contain constituents of Complexes I, III, IV, and V as well as accessory proteins involved in supercomplex assembly and stability, cristae architecture, carbohydrate and fat oxidation, and oxidant detoxification. Respiratory analysis of high molecular mass supercomplexes confirmed the presence of intact respirasomes, capable of transferring electrons from NADH to O2. Provision of respiratory substrates to isolated mitochondria augmented supercomplex abundance, with fatty acyl substrate (octanoylcarnitine) promoting higher supercomplex abundance than carbohydrate-derived substrate (pyruvate). Mitochondria isolated from transgenic hearts that express kinase-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (GlycoLo), which decreases glucose utilization and increases reliance on fatty acid oxidation for energy, had higher mitochondrial supercomplex abundance and activity compared with mitochondria from wild-type or phosphatase-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-expressing hearts (GlycoHi), the latter of which encourages reliance on glucose catabolism for energy. These findings indicate that high energetic reliance on fatty acid catabolism bolsters levels of mitochondrial supercomplexes, supporting the idea that the energetic state of the heart is regulatory factor in supercomplex assembly or stability.
DOI: 10.1016/j.freeradbiomed.2009.05.009
2009
Cited 33 times
Methods for imaging and detecting modification of proteins by reactive lipid species
Products of lipid peroxidation are generated in a wide range of pathologies associated with oxidative stress and inflammation. Many oxidized lipids contain reactive functional groups that can modify proteins, change their structure and function, and affect cell signaling. However, intracellular localization and protein adducts of reactive lipids have been difficult to detect, and the methods of detection rely largely on antibodies raised against specific lipid-protein adducts. As an alternative approach to monitoring oxidized lipids in cultured cells, we have tagged the lipid peroxidation substrate arachidonic acid and an electrophilic lipid, 15-deoxy-Delta(12,14)-prostaglandin-J2 (15d-PGJ2), with either biotin or the fluorophore BODIPY. Tagged arachidonic acid can be used in combination with conditions of oxidant stress or inflammation to assess the subcellular localization and protein modification by oxidized lipids generated in situ. Furthermore, we show that reactive lipid oxidation products such as 15d-PGJ2 can also be labeled and used in fluorescence and Western blotting applications. This article describes the synthesis, purification, and selected application of these tagged lipids in vitro.
DOI: 10.4236/ajps.2011.23051
2011
Cited 28 times
Assessing Influence of Ozone in Tomato Seed Dormancy Alleviation
The study was made on the role of ozone (O 3 ) gas treatment on seeds of Lycopersicon esculentum cv.PKM1 (tomato) to release dormancy in advance.The experimental conditions followed a complete factorial design with 3 independent factors, i.e.A faster start of germination in T 2 treatment (98% -99%) was observed than in other treatments (T 1 , T 3 and T 4 ) compared to control seeds by measuring seedling growth rate on 5 th day after treatment.Nevertheless a too long and high concentration of O 3 treatment seemed to be penalizing on germination rate whereas low O 3 concentration (T 2 ) for a moderate time interval (20 min) seemed to be most beneficial.The treated seeds were stored, checked 1, 3, 6 months later and found that, the seeds have retained their accelerated germination efficiency.In particular, 1T 2 (1 month storage), 3T 2 (3 month storage) had maximum germination rate among all the stored treated seeds, but 6T 2 (6 month storage) didn't show sustained germination acceleration efficiency.Therefore, it was found that when time prolongs, O 3 treatment loss its effect steadily and the germination efficiency of all the treatments are more or less same at six months after treatment.It is hypothesized that the application of O 3 acts as an important phenomenon in accelerating seed germination by breaking the dormancy in advance which is associated with reduced level of ABA in O 3 treated seeds.
DOI: 10.1042/bj20101390
2011
Cited 27 times
Responses of hypertrophied myocytes to reactive species: implications for glycolysis and electrophile metabolism
During cardiac remodelling, the heart generates higher levels of reactive species; yet an intermediate ‘compensatory’ stage of hypertrophy is associated with a greater ability to withstand oxidative stress. The mechanisms underlying this protected myocardial phenotype are poorly understood. We examined how a cellular model of hypertrophy deals with electrophilic insults, such as would occur upon ischaemia or in the failing heart. For this, we measured energetics in control and PE (phenylephrine)-treated NRCMs (neonatal rat cardiomyocytes) under basal conditions and when stressed with HNE (4-hydroxynonenal). PE treatment caused hypertrophy as indicated by augmented atrial natriuretic peptide and increased cellular protein content. Hypertrophied myocytes demonstrated a 2.5-fold increase in ATP-linked oxygen consumption and a robust augmentation of oligomycin-stimulated glycolytic flux and lactate production. Hypertrophied myocytes displayed a protected phenotype that was resistant to HNE-induced cell death and a unique bioenergetic response characterized by a delayed and abrogated rate of oxygen consumption and a 2-fold increase in glycolysis upon HNE exposure. This augmentation of glycolytic flux was not due to increased glucose uptake, suggesting that electrophile stress results in utilization of intracellular glycogen stores to support the increased energy demand. Hypertrophied myocytes also had an increased propensity to oxidize HNE to 4-hydroxynonenoic acid and sustained less protein damage due to acute HNE insults. Inhibition of aldehyde dehydrogenase resulted in bioenergetic collapse when myocytes were challenged with HNE. The integration of electrophile metabolism with glycolytic and mitochondrial energy production appears to be important for maintaining myocyte homoeostasis under conditions of increased oxidative stress.
DOI: 10.3389/fphys.2014.00453
2014
Cited 21 times
Impact of nutrient excess and endothelial nitric oxide synthase on the plasma metabolite profile in mice
An increase in calorie consumption is associated with the recent rise in obesity prevalence. However, our current understanding of the effects of nutrient excess on major metabolic pathways appears insufficient to develop safe and effective metabolic interventions to prevent obesity. Hence, we sought to identify systemic metabolic changes caused by nutrient excess and to determine how endothelial nitric oxide synthase (eNOS)—which has anti-obesogenic properties—affects systemic metabolism by measuring plasma metabolites. Wild-type (WT) and eNOS transgenic (eNOS-TG) mice were placed on low fat or high fat diets for six weeks, and plasma metabolites were measured using an unbiased metabolomic approach. High fat feeding in WT mice led to significant increases in fat mass, which was associated with significantly lower plasma levels of 1,5-anhydroglucitol, lysophospholipids, 3-dehydrocarnitine, and bile acids, as well as branched chain amino acids (BCAAs) and their metabolites. Plasma levels of several lipids including sphingomyelins, stearoylcarnitine, dihomo-linoleate and metabolites associated with oxidative stress were increased by high fat diet. In comparison with low fat-fed WT mice, eNOS-TG mice showed lower levels of several free fatty acids, but in contrast, the levels of bile acids, amino acids, and BCAA catabolites were increased. When placed on a high fat diet, eNOS overexpressing mice showed remarkably higher levels of plasma bile acids and elevated levels of plasma BCAAs and their catabolites compared with WT mice. Treatment with GW4064, an inhibitor of bile acid synthesis, decreased plasma bile acid levels but was not sufficient to reverse the anti-obesogenic effects of eNOS overexpression. These findings reveal unique metabolic changes in response to high fat diet and eNOS overexpression and suggest that the anti-obesity effects of eNOS are likely independent of changes in the bile acid pool.
DOI: 10.1039/c7ay00291b
2017
Cited 20 times
Analysis of stable isotope assisted metabolomics data acquired by high resolution mass spectrometry
Stable isotope assisted metabolomics (SIAM) uses stable isotope tracers to support studies of biochemical mechanisms. We report a suite of data analysis algorithms for automatic analysis of SIAM data acquired on a high resolution mass spectrometer. To increase the accuracy of isotopologue assignment, metabolites detected in the unlabeled samples were used as reference metabolites to generate possible isotopologue candidates for analysis of peaks detected in the labeled samples. An iterative linear regression model was developed to deconvolute the overlapping isotopic peaks of isotopologues present in a full MS spectrum, where the threshold for the weight factor was determined by a simulation study assuming different levels of Gaussian white noise contamination. A normalization method enabling isotope ratio-based normalization was implemented to study the difference of isotopologue abundance distribution between sample groups. The developed method can analyze SIAM data acquired by direct infusion MS and LC-MS, and can handle metabolite tracers containing different tracer elements. Analysis of SIAM data acquired from mixtures of known compounds showed that the developed algorithms accurately identify metabolites and quantify stable isotope enrichment. Application of SIAM data acquired from a biological study further demonstrated the effectiveness and accuracy of the developed method for analysis of complex samples.
DOI: 10.1038/s41598-019-50183-3
2019
Cited 19 times
Integration of flux measurements and pharmacological controls to optimize stable isotope-resolved metabolomics workflows and interpretation
Stable isotope-resolved metabolomics (SIRM) provides information regarding the relative activity of numerous metabolic pathways and the contribution of nutrients to specific metabolite pools; however, SIRM experiments can be difficult to execute, and data interpretation is challenging. Furthermore, standardization of analytical procedures and workflows remain significant obstacles for widespread reproducibility. Here, we demonstrate the workflow of a typical SIRM experiment and suggest experimental controls and measures of cross-validation that improve data interpretation. Inhibitors of glycolysis and oxidative phosphorylation as well as mitochondrial uncouplers serve as pharmacological controls, which help define metabolic flux configurations that occur under well-controlled metabolic states. We demonstrate how such controls and time course labeling experiments improve confidence in metabolite assignments as well as delineate metabolic pathway relationships. Moreover, we demonstrate how radiolabeled tracers and extracellular flux analyses integrate with SIRM to improve data interpretation. Collectively, these results show how integration of flux methodologies and use of pharmacological controls increase confidence in SIRM data and provide new biological insights.
DOI: 10.4049/jimmunol.1900144
2019
Cited 18 times
Exercise Promotes Resolution of Acute Inflammation by Catecholamine-Mediated Stimulation of Resolvin D1 Biosynthesis
Abstract The mechanisms by which regular exercise prevents the development and progression of chronic inflammatory diseases are largely unknown. We find that exercise enhances resolution of acute inflammation by augmenting resolvin D1 (RvD1) levels and by promoting macrophage phagocytosis. When compared with sedentary controls, mice that performed a four-week treadmill exercise regimen displayed higher macrophage phagocytic activity, enhanced RvD1 levels, and earlier neutrophil clearance following an acute inflammatory challenge. In acute inflammatory cell extracts from exercised mice, we found elevated expression of Alox15 and Alox5 and higher RvD1 levels. Because exercise stimulates release of epinephrine, which has immunomodulatory effects, we questioned whether epinephrine exerts proresolving actions on macrophages. Epinephrine-treated macrophages displayed higher RvD1 levels and 15-lipoxygenase-1 protein abundance, which were prevented by incubation with the α1 adrenergic receptor (α1-AR) antagonist prazosin. Likewise, stimulation of the α1-AR with phenylephrine enhanced macrophage phagocytosis and RvD1 production. During acute inflammation, prazosin abrogated exercise-enhanced neutrophil clearance, macrophage phagocytosis, and RvD1 biosynthesis. These results suggest that exercise-stimulated epinephrine enhances resolution of acute inflammation in an α1-AR–dependent manner. To our knowledge, our findings provide new mechanistic insights into the proresolving effects of exercise that could lead to the identification of novel pathways to stimulate resolution.
DOI: 10.1016/j.jprot.2017.03.014
2017
Cited 17 times
Systems characterization of differential plasma metabolome perturbations following thrombotic and non-thrombotic myocardial infarction
Myocardial infarction (MI) is an acute event characterized by myocardial necrosis. Thrombotic MI is caused by spontaneous atherosclerotic plaque disruption that results in a coronary thrombus; non-thrombotic MI occurs secondary to oxygen supply-demand mismatch. We sought to characterize the differential metabolic perturbations associated with these subtypes utilizing a systems approach. Subjects presenting with thrombotic MI, non-thrombotic MI and stable coronary artery disease (CAD) were included. Whole blood was collected at two acute time-points and at a time-point representing the quiescent stable disease state. Plasma metabolites were analyzed by untargeted UPLC-MS/MS and GC-MS. A weighted network was constructed, and modules were determined from the resulting topology. To determine perturbed modules, an enrichment analysis for metabolites that demonstrated between-group differences in temporal change across the disease state transition was then conducted.We report evidence of metabolic perturbations of acute MI and determine perturbations specific to thrombotic MI. Specifically, a module characterized by elevated glucocorticoid steroid metabolites following acute MI showed greatest perturbation following thrombotic MI. Modules characterized by elevated pregnenolone metabolites, monoacylglycerols, and acylcarnitines were perturbed following acute MI. A module characterized by a decrease in plasma amino acids following thrombotic MI was differentially perturbed between MI subtypes.
DOI: 10.18632/oncotarget.22575
2017
Cited 17 times
Distinct roles of TRAF6 and TAK1 in the regulation of adipocyte survival, thermogenesis program, and high-fat diet-induced obesity
Chronic low-grade inflammation, adipocyte hypertrophy, and glucose intolerance are common features of obesity and a risk factor for cancer.Tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6) is an adaptor protein that also possesses a non-conventional E3 ubiquitin ligase activity.In response to receptor-mediated events, TRAF6 activates transforming growth factor-activated kinase 1 (TAK1), which leads to activation of the MAPK and nuclear factor-kappa B (NF-κB) signaling pathways.However, the roles of TRAF6 and TAK1 in the regulation of adipocyte function remain less understood.Here, we demonstrate that adipocyte-specific deletion of TAK1, but not TRAF6, in mice reduces the survival of adipocytes and abundance of white adipose tissue (WAT).Adipocyte-specific ablation of TAK1, but not TRAF6, increases the expression for markers of beige/brown fat in WAT.Deletion of TAK1 in WAT increases phosphorylation of AMPK, abundance of PGC-1α, non-canonical NF-κB signaling, markers of M2 macrophages, and diminishes phosphorylation of JNK and canonical NF-κB signaling.Levels of TRAF6 and enzymatic activity of TAK1 are increased in WAT of mice fed with high-fat diet (HFD).Our results demonstrate that ablation of TAK1 drastically reduces HFD-induced obesity and improves energy expenditure and glucose metabolism.In contrast, adipocyte-specific ablation of TRAF6 has a minimal effect on HFD-induced obesity.Collectively, our results suggest that even though TRAF6 is an upstream activator of TAK1 in many signaling cascades, inactivation of TAK1, but not TRAF6, regulates adipocyte survival, energy expenditure, and HFD-induced obesity in mice.
DOI: 10.1016/j.redox.2019.101177
2019
Cited 17 times
Mitochondria-associated lactate dehydrogenase is not a biologically significant contributor to bioenergetic function in murine striated muscle
Previous studies indicate that mitochondria-localized lactate dehydrogenase (mLDH) might be a significant contributor to metabolism. In the heart, the presence of mLDH could provide cardiac mitochondria with a higher capacity to generate reducing equivalents directly available for respiration, especially during exercise when circulating lactate levels are high. The purpose of this study was to test the hypothesis that mLDH contributes to striated muscle bioenergetic function. Mitochondria isolated from murine cardiac and skeletal muscle lacked an appreciable ability to respire on lactate in the absence or presence of exogenous NAD+. Although three weeks of treadmill running promoted physiologic cardiac growth, mitochondria isolated from the hearts of acutely exercised or exercise-adapted mice showed no further increase in lactate oxidation capacity. In all conditions tested, cardiac mitochondria respired at >20-fold higher levels with provision of pyruvate compared with lactate. Similarly, skeletal muscle mitochondria showed little capacity to respire on lactate. Protease protection assays of isolated cardiac mitochondria confirmed that LDH is not localized within the mitochondrion. We conclude that mLDH does not contribute to cardiac bioenergetics in mice.
DOI: 10.1016/j.yjmcc.2021.08.013
2022
Cited 7 times
In vivo deep network tracing reveals phosphofructokinase-mediated coordination of biosynthetic pathway activity in the myocardium
Glucose metabolism comprises numerous amphibolic metabolites that provide precursors for not only the synthesis of cellular building blocks but also for ATP production. In this study, we tested how phosphofructokinase-1 (PFK1) activity controls the fate of glucose-derived carbon in murine hearts in vivo. PFK1 activity was regulated by cardiac-specific overexpression of kinase- or phosphatase-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase transgenes in mice (termed GlycoLo or GlycoHi mice, respectively). Dietary delivery of 13C6-glucose to these mice, followed by deep network metabolic tracing, revealed that low rates of PFK1 activity promote selective routing of glucose-derived carbon to the purine synthesis pathway to form 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR). Consistent with a mechanism of physical channeling, we found multimeric protein complexes that contained phosphoribosylaminoimidazole carboxylase (PAICS)-an enzyme important for AICAR biosynthesis, as well as chaperone proteins such as Hsp90 and other metabolic enzymes. We also observed that PFK1 influenced glucose-derived carbon deposition in glycogen, but did not affect hexosamine biosynthetic pathway activity. These studies demonstrate the utility of deep network tracing to identify metabolic channeling and changes in biosynthetic pathway activity in the heart in vivo and present new potential mechanisms by which metabolic branchpoint reactions modulate biosynthetic pathways.
DOI: 10.1016/j.molmet.2022.101637
2022
Cited 7 times
Exercise-induced specialized proresolving mediators stimulate AMPK phosphorylation to promote mitochondrial respiration in macrophages
Physical activity has been shown to reduce the risk of CVD mortality in large-cohort longitudinal studies; however, the mechanisms underpinning the beneficial effects of exercise remain incompletely understood. Emerging data suggest that the risk reducing effect of exercise extends beyond changes in traditional CVD risk factors alone and involves alterations in immunity and reductions in inflammatory mediator production. Our study aimed to determine whether exercise-enhanced production of proresolving lipid mediators contribute to alterations in macrophage intermediary metabolism, which may contribute to the anti-inflammatory effects of exercise.Changes in lipid mediators and macrophage metabolism were assessed in C57Bl/6 mice following 4 weeks of voluntary exercise training. To investigate whether exercise-stimulated upregulation of specialized proresolving lipid mediators (SPMs) was sufficient to enhance mitochondrial respiration, both macrophages from control mice and human donors were incubated in vitro with SPMs and mitochondrial respiratory parameters were measured using extracellular flux analysis. Compound-C, an ATP-competitive inhibitor of AMPK kinase activity, was used to investigate the role of AMPK activity in SPM-induced mitochondrial metabolism. To assess the in vivo contribution of 5-lipoxygenase in AMPK activation and exercise-induced mitochondrial metabolism in macrophages, Alox5-/- mice were also subjected to exercise training.Four weeks of exercise training enhanced proresolving lipid mediator production, while also stimulating the catabolism of inflammatory lipid mediators (e.g., leukotrienes and prostaglandins). This shift in lipid mediator balance following exercise was associated with increased macrophage mitochondrial metabolism. We also find that treating human and murine macrophages in vitro with proresolving lipid mediators enhances mitochondrial respiratory parameters. The proresolving lipid mediators RvD1, RvE1, and MaR1, but not RvD2, stimulated mitochondrial respiration through an AMPK-dependent signaling mechanism. Additionally, in a subset of macrophages, exercise-induced mitochondrial activity in vivo was dependent upon 5-lipoxygenase activity.Collectively, these results suggest that exercise stimulates proresolving lipid mediator biosynthesis and mitochondrial metabolism in macrophages via AMPK, which might contribute to the anti-inflammatory and CVD risk reducing effect of exercise.
DOI: 10.3390/diagnostics14070669
2024
Development of a Musculoskeletal Ultrasound Protocol to Evaluate Hand Pain in Systemic Sclerosis Patients
Hand impairment is a frequently reported complaint in systemic sclerosis (SSc) patients and a leading cause of disability and diminished quality of life. Managing hand pain can be particularly challenging due to the coexistence of non-inflammatory arthralgias, inflammatory arthritis, acro-osteolysis, tenosynovitis, joint contractures, tendon friction rubs, nerve entrapment, Raynaud's phenomenon (RP), digital ulcers (DU), sclerodactyly, calcinosis, and chronic pain. While physical examination and radiographs are the first line methods for evaluating hand pain, they are limited in scope and miss many underlying etiologies of hand impairment. We propose a joint ultrasound (US) hand protocol to differentiate between various articular, periarticular, ischemic, skin, and nerve pathologies and to assist in targeted treatment strategies.
DOI: 10.1152/ajpheart.00127.2024
2024
Cardiac Mitochondrial Metabolism During Pregnancy and the Postpartum Period
The goal of the present study was to characterize changes in mitochondrial respiration in the maternal heart during pregnancy and after birth. Timed pregnancy studies were performed in 12-week-old female FVB/NJ mice, and cardiac mitochondria were isolated from the following groups of mice: non-pregnant (NP), mid-pregnancy (MP), late-pregnancy (LP), and 1-week post-birth (PB). Similar to our previous studies, we observed increased heart size during all stages of pregnancy (e.g., MP and LP) and post-birth (e.g., PB) compared with NP mice. Differential cardiac gene and protein expression analyses revealed changes in several mitochondrial transcripts at LP and PB, including several mitochondrial complex subunits and members of the Slc family, important for mitochondrial substrate transport. Respirometry revealed that pyruvate- and glutamate-supported state 3 respiration was significantly higher in PB versus LP mitochondria, with respiratory control ratio (RCR) values higher in PB mitochondria. In addition, we found that PB mitochondria respired more avidly when given 3-hydroxybutyrate (3-OHB) than mitochondria from NP, MP, and LP hearts, with no differences in RCR. These increases in respiration in PB hearts occurred independent of changes in mitochondrial yield, but were associated with higher abundance of 3-hydroxybutyrate dehydrogenase 1. Collectively, these findings suggest that, after birth, maternal cardiac mitochondria have an increased capacity to use 3-OHB, pyruvate, and glutamate as energy sources; however, increases in mitochondrial efficiency in the postpartum heart appear limited to carbohydrate and amino acid metabolism.
DOI: 10.1016/j.yjmcc.2024.04.014
2024
Exercise-induced changes in myocardial glucose utilization during periods of active cardiac growth
Exercise training can promote physiological cardiac growth, which has been suggested to involve changes in glucose metabolism to facilitate hypertrophy of cardiomyocytes. In this study, we used a dietary, in vivo isotope labeling approach to examine how exercise training influences the metabolic fate of carbon derived from dietary glucose in the heart during acute, active, and established phases of exercise-induced cardiac growth. Male and female FVB/NJ mice were subjected to treadmill running for up to 4 weeks and cardiac growth was assessed by gravimetry. Cardiac metabolic responses to exercise were assessed via in vivo tracing of [
DOI: 10.1097/sap.0000000000003944
2024
Amputated Digit Replantations
Introduction A common consideration for replantation success is the ischemia time following injury and the preservation temperature. A classic principle within the hand surgery community describes 12 hours of warm ischemia and 24 hours of cold ischemia as the upper limits for digit replantation; however, these limits are largely anecdotal and based on older studies. We aimed to compare survival data from the large body of literature to aid surgeons and all those involved in the replantation process in hopes of optimizing success rates. Methods The PubMed database was queried on April 4th, 2023, for articles that included data on digit replantation survival in terms of temperature of preservation and ischemia time. All primary outcomes were analyzed with the Mantel-Haenszel method within a random effects model. Secondary outcomes were pooled and analyzed using the chi-square statistic. Statistical analysis and forest plot generation were completed with RevMan 5.4 software with odds ratios calculated within a 95% confidence interval. Results Our meta-analysis identified that digits preserved in cold ischemia for over 12 hours had significantly higher odds of replantation success than the amputated digits replanted with 0–12 hours of warm ischemia time ( P ≤ 0.05). The odds of survival in the early (0–6 hours) replantation group were around 40% greater than the later (6–12 hours) replantation group ( P ≤ 0.05). Secondary outcomes that were associated with higher survival rates included a clean-cut amputation, increased venous and arterial anastomosis, a repair that did not require a vein graft, and replants performed in nonsmokers ( P ≤ 0.05). Discussion Overall, these findings suggest that when predicting digit replantation success, time is of the essence when the digit has yet to be preserved in a cold environment. This benefit, however, is almost completely diminished when the amputated digit is appropriately maintained in a cold environment soon after injury. In conclusion, our results suggest that there is potential for broadening the ischemia time limits for digit replant survival outlined in the literature, particularly for digits that have been stored correctly in cold ischemia.
DOI: 10.1152/physiol.2024.39.s1.1619
2024
Lactate Suppresses, While Glutamine or Branched-Chain Amino Acids Support, Cardiac Fibroblast Activation
Background: Although metabolic changes occur during activation of cardiac fibroblasts, it remains unclear how different metabolic pathways influence fibroblast phenotype and function. Substrates used for biosynthetic pathways could be particularly important to the fibrotic response because activated fibroblasts require higher anabolic output to provide biomolecular building blocks for cell division and ECM secretion. Hence, elucidating how metabolism influences fibroblast differentiation and activation could provide valuable information for understanding mechanisms of fibrosis. Hypothesis: We hypothesized that the availability of specific metabolic substrates regulate myofibroblast differentiation. Methods and Results: Naïve cardiac fibroblasts were isolated from wildtype, male C57BL/6J mice and treated with TGFβ or bFGF for 48 h. To examine metabolic genes that respond to profibrotic stimuli, we utilized RNA sequencing, which indicated that TGFβ significantly increases transcription of numerous glycolytic and glutamine metabolism genes as well as transporters for branched-chain amino acids (BCAAs), glutamine, and lactate. Guided by these results, we used an αSMA-reporter assay to examine how lactate, glutamine and BCAA availability influence TGFβ-induced myofibroblast differentiation. Fibroblasts were treated with deficient media supplemented with increasing concentrations of substrate and challenged with TGFβ for 48 hours, followed by assessment of αSMA positivity. In a dose-dependent manner, fibroblasts treated with lactate exhibited decreased αSMA reporter activity (EC 50 =6.17mM). Furthermore, extracellular flux analysis revealed that the presence of lactate (1mM–10mM) significantly reduced basal oxygen consumption rate and extracellular acidification rate in TGFβ-treated cells (n=2, p&lt;0.05). In contrast, αSMA expression was blunted with diminishing concentrations of glutamine (n=3, p&lt;0.05) and BCAAs, indicating suffcient supply of these substrates is essential for fibroblast differentiation. In addition, immunoblotting data of TGFβ-treated fibroblasts revealed that low extracellular BCAA levels (0 or 100μM) decreased abundance of col1a1 and periostin compared with treatment with BCAAs within the physiological range (400μM; n=4, p&lt;0.05). Conclusion: These preliminary findings provide insight into how specific alterations of glycolysis, glutaminolysis, and BCAA metabolism modulate fibroblast activation in vitro. Additional studies that include alterations to other critical substrates (glucose, fatty acids, ketone bodies, etc.) and enzymes of ancillary biosynthetic pathways of glucose metabolism could provide additional insights into how metabolism regulates fibroblast function. Ultimately, the goal of these studies is to discover potential genetic and pharmacological interventions that positively influence wound healing in the infarcted heart and limit the burden of cardiac fibrosis. NIH and AHA. This is the full abstract presented at the American Physiology Summit 2024 meeting and is only available in HTML format. There are no additional versions or additional content available for this abstract. Physiology was not involved in the peer review process.
DOI: 10.1152/physiol.2024.39.s1.1670
2024
TAK1 Signaling in Cardiac Fibroblast Contributes to Inflammation Resolution Post Myocardial Infarction
Introduction: Resident cardiac fibroblasts are often defined by their extraordinary capacity to deposit extracellular matrix after cardiac injury; however, fibroblasts also react avidly to pro-inflammatory stimuli, suggesting that they may also be mediators of inflammation. Initially identified as a TGFβ responsive enzyme, TGF-β activated kinase 1 (TAK1) has been shown to be significant in regulating pro-inflammatory signaling in several disease models. The goal of this study was to understand how fibroblasts integrate pro-inflammatory signals to regulate acute inflammation following myocardial infarction (MI). We hypothesize that cardiac fibroblasts utilize a TAK1-dependent mechanism to integrate pro-inflammatory signals and orchestrate the cellular immune response following injury. Methods and Results: To create a fibroblast-specific model for TAK1 knockdown, TAK1 fl/fl mice were bred to Col1a2-Cre ERT mice. Following 2 weeks of tamoxifen chow feeding, both Cre − and Cre + TAK1 fl/fl mice underwent permanent coronary ligation to induce MI. Three days following MI, immune cell populations from the heart, spleen, and peripheral blood were evaluated through flow cytometry. Deletion of TAK1 in fibroblasts in vivo diminished neutrophils and Ly6C low patrolling monocytes populations in the heart and decreased the number of CCR2 neg resident macrophages after MI (n=10/group; p&lt;0.05). Interestingly, circulating levels of neutrophils in the peripheral blood as well as those in the spleen were not significantly different between the groups. To determine how TAK1 may regulate pro-inflammatory signaling, isolated cardiac fibroblasts from TAK1 fl/fl mice were treated with Ad-CMV-iCre and were subsequently treated with IL1β. Notably, TAK1 knockdown suppressed IL1β-dependent p-JNK and NFκB signaling as well as MMP3 production. Further LC-MS/MS evaluation of lipid mediators in conditioned medium showed that deletion of TAK1 significantly increased fibroblast-derived resolvins (RvE2, RvD1, RvD4, RvE4) and lipoxins (LXA 4 , 15( R)-LXA 4 ) (n= 3-5; p&lt;0.05). Conclusion: Resident cardiac fibroblasts regulate the acute immune response following MI via a TAK1-dependent mechanism. Secreted factors derived from cardiac fibroblasts likely play a pivotal role in shaping the inflammatory and immune response after MI. NIH and AHA. This is the full abstract presented at the American Physiology Summit 2024 meeting and is only available in HTML format. There are no additional versions or additional content available for this abstract. Physiology was not involved in the peer review process.
DOI: 10.1152/physiol.2024.39.s1.2408
2024
Cardiomyocyte Ketone Metabolism Regulates Myocardial Hyperemia
Myocardial perfusion is inextricably coupled to metabolism, with increases in physiologic cardiac demand promoting coronary vasodilation. Despite its wide acceptance, the hypothesis that cardiomyocyte metabolism autoregulates myocardial perfusion lacks mechanistic detail; however, clinical and preclinical studies suggest a signaling role for intermediary metabolites in the vasodilatory response. In particular, the ketone body precursor, β-hydroxybutyrate (3-OHB), enhances myocardial blood flow in humans, but the mechanism by which it does so remains unclear. Here, we determined the extent to which 3-OHB influences myocardial hyperemia in mice. Intravenous 3-OHB infusion (0.5 g/kg) in adult mice (12–24 wks) raised circulating 3-OHB levels (0.5 ± 0.1 to 5.1 ± 0.6 mM; p=0.005) and increased myocardial perfusion (18.6 ± 4.4 to 50.9 ± 10.4 mL/min/g; p=0.024), as measured by myocardial contrast echocardiography. Enhanced perfusion induced by 3-OHB occurred in the absence of changes in cardiac output or systolic performance. Next, to determine if 3-OHB directly dilates the coronary vasculature, we exposed pressurized (80 mmHg), small-diameter (100–260 μm) coronary arteries from mice (C57/Bl6J and 6N) to 3-OHB (0.1–5 mM) but found no significant changes in diameter. Based on these results, we tested whether cardiomyocyte metabolism of 3-OHB is required for the vasodilatory response. We deleted β-hydroxybutyrate dehydrogenase (BDH1) selectively in cardiomyocytes using Bdh1 fl/fl mice crossed with tamoxifen-inducible Cre recombinase mice (csBDH1 −/− ). Although 3-OHB robustly enhanced myocardial perfusion (6.9 ± 1.4 to 21.3 ± 5.6 mL/min/g; p=0.031) at sustained cardiac workloads and peripheral vascular resistance in Cre-negative littermate (control) mice, csBDH1 −/− mice completely lacked the 3-OHB-induced hyperemic response (8.0 ± 2.5 to 6.3 ± 2.8 mL/min/g; p&gt;0.999). Together, these data indicate that 3-OHB evokes workload-independent enhancement of myocardial perfusion through a mechanism that requires cardiomyocyte metabolism of 3-OHB. Our results support the general concept that acute changes in metabolic substrate availability may influence oxygen delivery to cardiomyocytes via a cardiomyocyte-vascular signaling nexus. R01HL163818, R01HL142710, R01HL163003, P30GM127607, P30GM127607-05S1, Jewish Heritage Fund for Excellence (JHFE), S10OD025178. This is the full abstract presented at the American Physiology Summit 2024 meeting and is only available in HTML format. There are no additional versions or additional content available for this abstract. Physiology was not involved in the peer review process.
DOI: 10.1152/physiol.2024.39.s1.1744
2024
Neutrophil NADPH oxidase regulation by oxidative phosphorylation and accessory glucose metabolic pathways
Neutrophils produce reactive oxygen species (ROS) to extinguish pathogens and regulate intracellular signaling. The major producer of ROS in neutrophils is the NADPH oxidase, which is fueled by NADPH generated via the oxidative pentose phosphate pathway. It remains unclear how other accessory pathways of glucose metabolism and mitochondrial activity influence the oxidative burst in neutrophils. The respiratory burst in bone marrow-derived neutrophils was stimulated with phorbol 12-myristate 13-acetate (PMA) in the presence or absence of an inhibitor of glycogen breakdown (glycogen phosphorylase inhibitor; GPi), inhibitors of mitochondrial respiration (antimycin A and rotenone), or an inhibitor of serine biosynthesis (NCT503). Respiratory burst was also measured under conditions of limited glucose and glutamine. ROS production was measured via extracellular flux analysis or via dihydrorhodamine fluorescence. In both male and female mouse neutrophils, inhibition of glycogen breakdown by GPi delayed initiation of the oxidative burst in response to PMA (n = 6, p&lt;0.05, Student’s T-test and Repeated Measure’s ANOVA). Inhibition of mitochondrial oxidative phosphorylation or inhibition of the serine biosynthetic pathway sustained ROS production in PMA-stimulated neutrophils (n = 6, p&lt;0.05, Student’s T-test). Removal of glucose and glutamine showed a truncated oxidative burst (n = 6, p&lt;0.05, Student’s T-test). Ultimately, these findings suggest that glycogen breakdown fuels the initial activation of the oxidative burst, while mitochondrial activity and the serine biosynthesis pathway control the magnitude and duration of neutrophil ROS production. Furthermore, utilization of exogenous glucose and glutamine are necessary for the entire oxidative burst. Future studies will continue to illustrate the mechanisms of neutrophil metabolic plasticity as well as connect their significance in the context of wound healing, exercise, and chronic inflammatory diseases. NIH: R01HL141191, NIH: P01 HL078825. This is the full abstract presented at the American Physiology Summit 2024 meeting and is only available in HTML format. There are no additional versions or additional content available for this abstract. Physiology was not involved in the peer review process.
DOI: 10.1016/j.freeradbiomed.2012.08.014
2013
Cited 18 times
Utilization of fluorescent probes for the quantification and identification of subcellular proteomes and biological processes regulated by lipid peroxidation products
Oxidative modifications to cellular proteins are critical in mediating redox-sensitive processes such as autophagy, the antioxidant response, and apoptosis. The proteins that become modified by reactive species are often compartmentalized to specific organelles or regions of the cell. Here, we detail protocols for identifying the subcellular protein targets of lipid oxidation and for linking protein modifications with biological responses such as autophagy. Fluorophores such as BODIPY-labeled arachidonic acid or BODIPY-conjugated electrophiles can be paired with organelle-specific probes to identify specific biological processes and signaling pathways activated in response to oxidative stress. In particular, we demonstrate "negative" and "positive" labeling methods using BODIPY-tagged reagents for examining oxidative modifications to protein nucleophiles. The protocol describes the use of these probes in slot immunoblotting, quantitative Western blotting, in-gel fluorescence, and confocal microscopy techniques. In particular, the use of the BODIPY fluorophore with organelle- or biological process-specific dyes and chromophores is highlighted. These methods can be used in multiple cell types as well as isolated organelles to interrogate the role of oxidative modifications in regulating biological responses to oxidative stress.
DOI: 10.1016/b978-0-12-800254-4.00013-1
2014
Cited 16 times
Antiobesogenic Role of Endothelial Nitric Oxide Synthase
The prevalence of obesity has increased remarkably in the past four decades. Because obesity can promote the development of type 2 diabetes and cardiovascular disease, understanding the mechanisms that engender weight gain and discovering safe antiobesity therapies are of critical importance. In particular, the gaseous signaling molecule, nitric oxide (NO), appears to be a central factor regulating adiposity and systemic metabolism. Obese and diabetic states are characterized by a deficit in bioavailable NO, with such decreases commonly attributed to downregulation of endothelial NO synthase (eNOS), loss of eNOS activity, or quenching of NO by its reaction with oxygen radicals. Gain-of-function studies, in which vascular-derived NO has been increased pharmacologically or genetically, reveal remarkable actions of NO on body composition and systemic metabolism. This review addresses the metabolic actions of eNOS and the potential therapeutic utility of harnessing its antiobesogenic effects.
DOI: 10.1042/bj20080716
2008
Cited 20 times
S-Nitrosation and thiol switching in the mitochondrion: a new paradigm for cardioprotection in ischaemic preconditioning
Understanding the molecular mechanisms through which the heart could be protected from ischaemic injury is of major interest and offers a potential route for the development of new therapies. Recently, several studies have uncovered intriguing relationships between nitric oxide-induced protein thiol modifications and the cardioprotected phenotype. In a highly cited, seminal article published in the Biochemical Journal in 2006, Burwell and colleagues addressed this issue and provided direct evidence for S-nitrosation of complex I of the mitochondrial electron transport chain. These authors were the first to show increased S-nitrosation of mitochondrial proteins from hearts subjected to the cardioprotective process known as ischaemic preconditioning. This study has paved the way for further investigations that collectively reveal a potential link between the mitochondrial S-nitrosoproteome and ischaemic preconditioning.
DOI: 10.1152/ajpendo.00345.2017
2018
Cited 14 times
Glutathione<i>S</i>-transferase P deficiency induces glucose intolerance via JNK-dependent enhancement of hepatic gluconeogenesis
Hepatic glutathione S-transferases (GSTs) are dysregulated in human obesity, nonalcoholic fatty liver disease, and diabetes. The multifunctional GST pi-isoform (GSTP) catalyzes the conjugation of glutathione with acrolein and inhibits c-Jun NH2-terminal kinase (JNK) activation. Herein, we tested whether GSTP deficiency disturbs glucose homeostasis in mice. Hepatic GST proteins were downregulated by short-term high-fat diet in wild-type (WT) mice concomitant with increased glucose intolerance, JNK activation, and cytokine mRNAs in the liver. Genetic deletion of GSTP did not affect body composition, fasting blood glucose levels, or insulin levels in mice maintained on a normal chow diet; however, compared with WT mice, the GSTP-null mice were glucose intolerant. In GSTP-null mice, pyruvate intolerance, reflecting increased hepatic gluconeogenesis, was accompanied by elevated levels of activated JNK, cytokine mRNAs, and glucose-6-phosphatase proteins in the liver. Treatment of GSTP-null mice with the JNK inhibitor 1,9-pyrazoloanthrone (SP600125) significantly attenuated pyruvate-induced hepatic gluconeogenesis and significantly altered correlations between hepatic cytokine mRNAs and metabolic outcomes in GSTP-null mice. Collectively, these findings suggest that hepatic GSTP plays a pivotal role in glucose handling by regulating JNK-dependent control of hepatic gluconeogenesis. Thus, hepatic GSTP-JNK dysregulation may be a target of new therapeutic interventions during early stages of glucose intolerance to prevent the worsening metabolic derangements associated with human obesity and its relentless progression to diabetes.
DOI: 10.1016/j.yjmcc.2020.12.007
2021
Cited 9 times
Considerations for using isolated cell systems to understand cardiac metabolism and biology
Changes in myocardial metabolic activity are fundamentally linked to cardiac health and remodeling. Primary cardiomyocytes, induced pluripotent stem cell-derived cardiomyocytes, and transformed cardiomyocyte cell lines are common models used to understand how (patho)physiological conditions or stimuli contribute to changes in cardiac metabolism. These cell models are helpful also for defining metabolic mechanisms of cardiac dysfunction and remodeling. Although technical advances have improved our capacity to measure cardiomyocyte metabolism, there is often heterogeneity in metabolic assay protocols and cell models, which could hinder data interpretation and discernment of the mechanisms of cardiac (patho)physiology. In this review, we discuss considerations for integrating cardiomyocyte cell models with techniques that have become relatively common in the field, such as respirometry and extracellular flux analysis. Furthermore, we provide overviews of metabolic assays that complement XF analyses and that provide information on not only catabolic pathway activity, but biosynthetic pathway activity and redox status as well. Cultivating a more widespread understanding of the advantages and limitations of metabolic measurements in cardiomyocyte cell models will continue to be essential for the development of coherent metabolic mechanisms of cardiac health and pathophysiology.