Palmitate-induced cardiac apoptosis is mediated through CPT-1 but not influenced by glucose and insulin

2002 ◽  
Vol 282 (2) ◽  
pp. H717-H725 ◽  
Author(s):  
Jennifer Y. Kong ◽  
Simon W. Rabkin

To test the hypothesis that regulation of palmitate metabolism, through carnitine palmitoyl transferase-1 (CPT-1) or through alterations of glycolysis, was involved in the pathway of palmitate-mediated cell death, cardiomyocytes were cultured from 7-day-old chick embryos. Palmitate-induced cell death, assessed by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay, was enhanced by carnitine, a cofactor needed for palmitate transport into mitochondria via CPT-1. Carnitine co-incubation with palmitate significantly ( P < 0.01) increased the amount of apoptotic cells, assessed by propidium iodine staining and fluorescent-activated cell sorting analysis compared with treatment with either palmitate or carnitine alone. The CPT-1 inhibitor oxfenicine significantly ( P < 0.05) blocked the cell death induced by the combination of palmitate and carnitine. The short-chain saturated fatty acid capric acid (100 μM), which is not likely transported by CPT-1, did not significantly affect cell viability, whereas the C18 saturated fatty acid stearic (100 μM) significantly ( P < 0.01) reduced cell viability and to a similar extent as palmitate. In contrast, there was no significant alteration of palmitate-induced cell death by cotreatment with 100 nM insulin + 2 g/l glucose or 1 mM lactate, which promote ATP generation by glycolysis rather than fatty acid oxidation. Fumonisin did not alter palmitate-induced cell death or apoptosis, suggesting that the effect of palmitate was not operative through increased ceramide synthesis. These results suggest that oxidation of palmitate through CPT-1 is involved in the production of apoptosis in cardiomyocytes.

2019 ◽  
pp. 11-20
Author(s):  
David P. Basile ◽  
Babu J. Padanilam

Acute kidney injury represents a significant clinical disorder associated with a rapid loss of renal function following a variety of potential insults. This chapter reviews multiple issues related to the pathophysiology of AKI with an emphasis on studies from animal models. Early responses following kidney injury include impaired hemodynamic and bioenergetic responses. Reductions in renal ATP levels occur as a result of compromised fatty acid oxidation and impaired compensation by glycolysis. Sustained reductions in perfusion contribute to extension of AKI characterized by complex inflammatory and cellular injury responses, often leading to cell death. Concurrently, the kidney displays an elegant repair response, leading to successful recovery in most cases, characterized in part by epithelial cell growth, while maladaptive or incomplete recovery of tubules or capillaries can predispose the development of interstitial fibrosis and CKD progression.


1975 ◽  
Vol 5 (4) ◽  
pp. 515-522 ◽  
Author(s):  
George S. Puritch

Fatty acids and their potassium soaps were screened for their toxicity to different life stages and eggs of the balsam woolly aphid (Adelgespiceae (Ratz.)). The most effective fatty acids for causing aphid mortality were in two major groups, one centering around capric acid (C10) within the low-chain saturated fatty acid series and the other around oleic acid (C18:1), within the unsaturated 18-carbon fatty acids. The potassium soaps were better aphicides than the corresponding acids; the soaps of caprylic, capric, oleic, and linoleic acids were the most effective. Eggs were less sensitive to the soaps than later stages of the aphid, and there was a large variation in their response to the soap treatments. The possibility of using fatty acids and soaps as a control for the balsam woolly aphid is discussed.


2016 ◽  
Vol 2016 ◽  
pp. 1-10 ◽  
Author(s):  
Vidula T. Vachharajani ◽  
Tiefu Liu ◽  
Xianfeng Wang ◽  
Jason J. Hoth ◽  
Barbara K. Yoza ◽  
...  

Sirtuins (SIRT), first discovered in yeast as NAD+ dependent epigenetic and metabolic regulators, have comparable activities in human physiology and disease. Mounting evidence supports that the seven-member mammalian sirtuin family (SIRT1–7) guard homeostasis by sensing bioenergy needs and responding by making alterations in the cell nutrients. Sirtuins play a critical role in restoring homeostasis during stress responses. Inflammation is designed to “defend and mend” against the invading organisms. Emerging evidence supports that metabolism and bioenergy reprogramming direct the sequential course of inflammation; failure of homeostasis retrieval results in many chronic and acute inflammatory diseases. Anabolic glycolysis quickly induced (compared to oxidative phosphorylation) for ROS and ATP generation is needed for immune activation to “defend” against invading microorganisms. Lipolysis/fatty acid oxidation, essential for cellular protection/hibernation and cell survival in order to “mend,” leads to immune repression. Acute/chronic inflammations are linked to altered glycolysis and fatty acid oxidation, at least in part, by NAD+ dependent function of sirtuins. Therapeutically targeting sirtuins may provide a new class of inflammation and immune regulators. This review discusses how sirtuins integrate metabolism, bioenergetics, and immunity during inflammation and how sirtuin-directed treatment improves outcome in chronic inflammatory diseases and in the extreme stress response of sepsis.


1993 ◽  
Vol 265 (5) ◽  
pp. E777-E782
Author(s):  
I. Sussman ◽  
V. Schultz ◽  
S. Gupta ◽  
C. Grady ◽  
N. B. Ruderman ◽  
...  

Isolated bovine cerebral microvessels (ICMV) were incubated with different metabolic fuels to determine their ability to support microvessel Na(+)-K(+)-ATPase (quantitated as ouabain-sensitive 86Rb+ uptake) and the ATP/ADP ratio. In comparison with ICMV incubated with glucose, Na(+)-K(+)-ATPase activity was reduced by 55% after a 3-h incubation in fuel-free medium and by 30-40% after incubation with beta-hydroxybutyrate, acetoacetate, or glutamate. However, Na(+)-K(+)-ATPase activity was not significantly decreased in ICMV incubated with pyruvate or oleate plus carnitine. In contrast, only glucose was able to maintain the ATP/ADP ratio. To evaluate the effect of endogenous fatty acid metabolism on these parameters, ICMV were incubated with bromostearate, an inhibitor of fatty acid oxidation. Bromostearate decreased both Na(+)-K(+)-ATPase activity and the ATP/ADP ratio, even in the presence of glucose. These results indicate that the varying effects of different fuels on Na(+)-K(+)-ATPase in ICMV cannot be explained solely by their effects on the ATP/ADP ratio or on glycolytic ATP generation. They suggest that other fuel-modulated factors play a key role in regulating this enzyme.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3922-3922
Author(s):  
Matthew Tcheng ◽  
Alessia Roma ◽  
Nawaz Ahmed ◽  
Richard Smith ◽  
Preethi Jayanth ◽  
...  

Targeting cell metabolism has emerged as a viable treatment strategy in acute myeloid leukemia (AML), a malignant hematological disease characterized by poor patient outcomes and limited chemotherapeutic options. Compared to the normal hematopoietic population, leukemia cells exhibit an altered phenotype characterized by increased mitochondrial mass as well as a greater reliance on oxidative phosphorylation and fatty acid oxidation (FAO) for survival. Mitochondrial FAO is a four-reaction process that catabolizes fatty acids to acetyl-CoA, generating reductive equivalents for the electron transport chain (ETC) and anaplerotic intermediates for the TCA cycle. Clinically approved FAO inhibitors, such as trimetazidine and ranolazine, are tissue-specific, targeting FAO in some tissues but affecting additional pathways in others. To better understand the potential clinical utility of targeting FAO, we systematically tested a panel of clinical and pre-clinical FAO inhibitors, reasoning that the most potent FAO inhibitor would lead to a novel anti-AML target. Avocadyne was the most potent anti-AML compound, inducing leukemic cell death (EC50: 2.5 µM) and suppressing clonogenic growth of primary samples, while sparing normal hematopoietic cells. Further, avocadyne (100mg/kg twice weekly for 5 weeks) reduced patient-derived AML cell engraftment in the bone marrow of immune deficient mice. As a component of avocatin-B, a mixture of two fatty alcohols previously determined to accumulate in the mitochondria, we confirmed that avocadyne inhibited long chain FAO using radiolabeled studies and high resolution respirometry. To identify a molecular target, avocadyne treated cells were immunoprecipitated with antibodies against each intramitochondrial enzyme involved in long chain FAO (e.g., very long acyl-CoA dehydrogenase (VLCAD; step 1) and the alpha and beta subunits of the mitochondrial trifunctional protein: HADHA, HADHB; steps 2-4). Immunoblotting and LC/MS analysis confirmed that avocadyne co-eluted with VLCAD, but not with HADHA or HADHB, confirming a direct physical interaction between avocadyne and VLCAD. VLCAD introduces a double bond to a fully saturated long chain fatty acid and reduces electron transfer flavoprotein (ETF)-bound FAD, transferring these electrons to the ETC. Using fluorescence spectrophotometry and respirometry, avocadyne directly inhibited VLCAD activity, resulting in reduced ETF-supported respiration. The activity of MCAD, an acyl-CoA dehydrogenase catalyzing medium chain fats, was not affected, suggesting avocadyne inhibits long chain FAO exclusively. Further, profiling of acyl-carnitines following avocadyne treatment also showed a pattern characteristic of long chain FAO inhibition at VLCAD. To further understand how VLCAD modulated avocadyne sensitivity, avocadyne-resistant and VLCAD knockdown cells were generated. Lentiviral knockdown of VLCAD sensitized leukemic cells to avocadyne-induced FAO inhibition and death. In contrast, cells resistant to avocadyne had increased protein expression of VLCAD but no change in other long chain FAO enzymes. With increased VLCAD-supported ETF respiration, higher concentrations of avocadyne were required to induce cell death, compared to the parental line. These results show that VLCAD expression modulated leukemic sensitivity to avocadyne. Inhibiting FAO at VLCAD triggered an adaptive metabolic switch towards glycolysis, characterized by increased extracellular acidification. This compensatory increase in glycolysis was ultimately insufficient to prevent the depletion of TCA metabolites and ATP, leading to leukemic death. In contrast, following avocadyne treatment, normal umbilical cord blood-derived mononuclear cells increased glycolytic as well as pyruvate dehydrogenase activity and had no decrease in ATP levels, cell viability, or clonogenic growth. Together, these results highlight, for the first time, VLCAD as a novel anti-AML target and further suggest the clinical utility of FAO inhibition as a potential anti-AML strategy. Disclosures Minden: Trillium Therapetuics: Other: licensing agreement. Schimmer:Medivir Pharmaceuticals: Research Funding; Jazz Pharmaceuticals: Consultancy; Novartis Pharmaceuticals: Consultancy; Otsuka Pharmaceuticals: Consultancy.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3947-3947
Author(s):  
Yoko Tabe ◽  
Kazumasa Sekihara ◽  
Kaori Saitoh ◽  
Vivian Ruvolo ◽  
Takashi Miida ◽  
...  

Abstract Adipocytes are the prevalent stromal cell type in adult bone marrow (BM), comprising approximately 60% of BM space in a 65-year old person. In BM environment, leukemia cells continuously adapt to deficient supply of nutrients and oxygen, acquiring quiescent and chemoresistant profiles. Fatty acid metabolism is one of the key energy pathways for AML survival (Samudio, J Clin Invest. 2010), and we previously demonstrated that AML cells activate oxidative phosphorylation and fatty acid oxidation (FAO) in the presence of BM-adipocytes (Tabe ASH 2015). These findings indicate the importance of FAO for AML cells survival under the adipocyte-abundant BM-microenvironment. A novel FAO inhibitor avocatinB, an odd-numbered carbon lipid derived from avocado fruit, has been recently shown to induce apoptosis and cell growth inhibition in AML cells (Lee, Cancer Res. 2015). In the present study, we investigated the molecular mechanisms of anti-leukemic effect of avocatinB in AML cells, utilizing THP1, OCI-AML3 and U937 AML cell lines co-cultured with human mesenchymal stem cells (MSC)-derived BM-adipocytes, mimicking the aging BM microenvironment. Treatment with avocatin B significantly induced ROS accumulation in U937 cells co-cultured with BM-adipocytes (MFI of ROS-sensitive dye; avocatinB (-) / (+); 164±50 / 581±85, p=0.04), whereas only minimum increase of ROS was observed in the absence of BM-adipocyte, indicating that avocatinB causes progressive oxidative damage in AML cells under the BM-adipocyte co-culture conditions. Of importance, avocatinB synergistically enhanced apoptotic effects of AraC in the presence of BM-adipocytes (combination index CI; adipocyte (-) / (+); THP1: 1.2 / 0.4, OCI-AML3: 0.7 / 0.3). Immunoblot analysis demonstrated that avocatinB activated the stress response kinase AMPK in THP1 and OCI-AML3 cells under BM-adipocyte co-culture conditions. AMPK is a crucial cellular energy sensor that regulates energy metabolism including FAO and gene transcription through mTOR inhibition. We therefore investigated the role of AMPK in avocatinB induced anti-leukemic effects on AML cells, utilizing AMPK knockdown (shAMPK) OCI-AML3 cells. shAMPK OCI-AML3 cells were significantly less sensitive to nutrient starvation-induced cell death in the absence of BM-adipocyte (p<0.01). While co-culture with BM-adipocytes protected control (nsAMPK) OCI-AML3 cells from spontaneous cell death, co-culture facilitated cell death of shAMPK cells. In turn, shAMPK OCI-AML cells were less sensitive to avocatinB compared to nsAMPKcells in the absence of BM-adipocyte with no additive/synergistic anti-proliferative effects of avocainB and AraC combination irrespective of the presence of BM-adipocytes (CI > 1.0). In nsAMPK cells, but not in shAMPKcells BM-adipocyte co-culture upregulated p-4EBP1 and cMyc expression which was abrogated by avocatinB and AraC combination treatment accompanied by induction of cleaved caspase 3. In summary, FAO inhibitor avocatinB induces pro-apoptotic effects through AMPK-dependent inhibition of mTOR signaling that disrupts energy homeostasis and induces ROS accumulation in AML cells under BM-adipocyte co-culture conditions. The ability of avocatinB to selectively enhance anti-leukemic effects of AraC in the presence of BM-adipocytes suggests that the strategies targeting FAO warrant further exploration in elderly AML patients. Disclosures Konopleva: Reata Pharmaceuticals: Equity Ownership; Abbvie: Consultancy, Research Funding; Genentech: Consultancy, Research Funding; Stemline: Consultancy, Research Funding; Eli Lilly: Research Funding; Cellectis: Research Funding; Calithera: Research Funding.


2021 ◽  
Author(s):  
Baichun Xing ◽  
Linlin Yang ◽  
Yanan Cui

Abstract Background Lidocaine is a local anesthetic that wildly used in surgical treatment and postoperative medical care for lung cancers. We hypothesized that lidocaine at clinical plasma concentration can inhibit CXCL12/CXCR4 axis-regulated cytoskeletal remodeling thereby reduce the migration of Non-small-cell lung cancers (NSCLC) cells. Methods We determined the effect of lidocaine at clinical plasma concentration on CXCL12-induced cell viability, apoptosis, cell death, monolayer cell wound healing rate, individual cell migration indicators, expression of CXCR4, CD44, and ICAM-1, intracellular Ca2+ level, and filamentous actin level alteration of NSCLC cells A549 and CXCR4-knocked down A549 cells using CCK-8, Bcl-2 ELISA, Cell death ELISA, wound healing assay, chemotaxis assay, western blotting, QPCR, Fura-2-based intracellular Ca2+ assay, and Fluorescein Phalloidin staining respectively. Results Lidocaine did not affect cell viability, apoptosis, and cell death but inhibited CXCL12-induced migration, intracellular Ca2+ releasing, and filamentous actin increase. Lidocaine decreased expression of CXCR4, increased CD44, but had no effect on ICAM-1. CXCL12 induced the increase of CD44 and ICAM-1 but did not affect CD44 in the presence of lidocaine. The knockdown of CXCR4 eliminated all the effects of lidocaine. The overexpression of CXCR4 promoted migration but the migration was inhibited by lidocaine. Conclusion Lidocaine at clinical plasma concentrations inhibited CXCL12-induced CXCR4 activation, thereby reduced the intracellular Ca2+-dependent cytoskeleton remodeling, resulting in slower migration of A549 cells.


Sign in / Sign up

Export Citation Format

Share Document