Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell Mol Gastroenterol Hepatol ; 17(2): 279-291, 2024.
Article in English | MEDLINE | ID: mdl-37844795

ABSTRACT

BACKGROUND & AIMS: Nonalcoholic fatty liver disease (NAFLD), and its more severe form, nonalcoholic steatohepatitis (NASH), is the leading cause for liver failure and liver cancer. Although the etiology is likely multifactorial, genes involved in regulating lipid metabolism are enriched in human NAFLD genome-wide association studies (GWAS), pointing to dysregulated lipid metabolism as a major pathogenic factor. Glycerol-3-phosphate acyltransferase 1 (GPAT1), encoded by GPAM, converts acyl-CoAs and glycerol-3-phosphate into lysophosphatidic acid and has been shown to regulate lipid accumulation in the liver. However, its role in mediating the progression from NAFLD to NASH has not been explored. METHODS: GPAT1-deficient mice were generated and challenged with diets inducing hepatic steatosis and NASH. Effects of GPAT1 deficiency on lipid and systemic metabolic end points were evaluated. RESULTS: Ablating GPAT1 globally or specifically in mouse hepatocytes reduced hepatic steatosis in the context of diet-induced or genetic obesity. Interestingly, blunting of progression from NAFLD to NASH in global GPAT1 knockout (KO) mice was model dependent. GPAT1 KO mice were protected from choline deficient, amino acid defined high-fat diet-induced NASH development, but not from the high fat, high carbohydrate, and high cholesterol diet-induced NASH. CONCLUSIONS: Our preclinical data support the notion that lipid metabolism pathways regulated by GPAT1 in hepatocytes play an essential role in NASH progression, albeit in a model-dependent manner.


Subject(s)
Non-alcoholic Fatty Liver Disease , Mice , Animals , Humans , Non-alcoholic Fatty Liver Disease/pathology , Genome-Wide Association Study , Glycerol , Diet, High-Fat/adverse effects , Mice, Knockout , Phosphates , Lipids
2.
Metabolites ; 10(11)2020 Nov 20.
Article in English | MEDLINE | ID: mdl-33233825

ABSTRACT

Stable isotope tracers can be used to quantify the activity of metabolic pathways. Specifically, 2H-water is quite versatile, and its incorporation into various products can enable measurements of carbohydrate, lipid, protein and nucleic acid kinetics. However, since there are limits on how much 2H-water can be administered and since some metabolic processes may be slow, it is possible that one may be challenged with measuring small changes in isotopic enrichment. We demonstrate an advantage of the isotope fractionation that occurs during gas chromatography, namely, setting tightly bounded integration regions yields a powerful approach for determining isotope ratios. We determined how the degree of isotope fractionation, chromatographic peak width and mass spectrometer dwell time can increase the apparent isotope labeling. Relatively simple changes in the logic surrounding data acquisition and processing can enhance gas chromatography-mass spectrometry measures of low levels of 2H-labeling, this is especially useful when asymmetrical peaks are recorded at low signal:background. Although we have largely focused attention on alanine (which is of interest in studies of protein synthesis), it should be possible to extend the concepts to other analytes and/or hardware configurations.

3.
ACS Chem Biol ; 15(10): 2623-2635, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32930572

ABSTRACT

The ability to target specific proteins for degradation may open a new door toward developing therapeutics. Although effort in chemistry is essential for advancing this modality, i.e., one needs to generate proteolysis targeting chimeras (bifunctional molecules, also referred to as PROTACS) or "molecular glues" to accelerate protein degradation, we suspect that investigations could also benefit by directing attention toward physiological regulation surrounding protein homeostasis, including the methods that can be used to examine changes in protein kinetics. This perspective will first consider some metabolic scenarios that might be of importance when one aims to change protein abundance by increasing protein degradation. Specifically, could protein turnover impact the apparent outcome? We will then outline how to study protein dynamics by coupling stable isotope tracer methods with mass spectrometry-based detection; since the experimental conditions could have a dramatic effect on protein turnover, special attention is directed toward the application of methods for quantifying protein kinetics using in vitro and in vivo models. Our goal is to present key concepts that should enable mechanistically informed studies which test targeted protein degradation strategies.


Subject(s)
Protein Biosynthesis/physiology , Proteins/analysis , Proteins/metabolism , Proteolysis/drug effects , Proteostasis/physiology , Animals , Humans , Isotope Labeling , Kinetics , Mass Spectrometry , Proteins/chemistry
4.
ACS Chem Biol ; 15(7): 1892-1900, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32396332

ABSTRACT

Lipid oxidation and biosynthesis are crucial for cell survival, especially for rapidly proliferating cancer cells in a heterogeneous metabolic environment. The storage of high-energy lipid reservoirs competitively advantages the cancer cell over non-neoplastic tissue. Disrupting lipid biosynthetic processes, through modulation of fatty acid (FA) esterification or de novo lipogenesis (DNL), is of interest in drug discovery. Mimicking the in vivo environment in vitro is also vital for testing the efficacy of potential drug compounds. We present here a stable isotope tracer-based approach for examining the impact of exogenous FA and oxygen tension on the pathways that affect lipid biosynthesis, including the rates of metabolic flux. By applying tandem mass spectrometry (MS/MS) analyses to studies using parallel tracers, we characterized the impact of FA bioavailability on the positional enrichment within specific lipids. Our observations suggest that adding bioavailable FA as a carbon source preferentially biases the cellular metabolism away from DNL and toward esterification of free fatty acid pools. Additionally, we have found that this FA addition, under hypoxic conditions, led to a biased increase in the total triglyceride pool (nearly 5-fold, as compared to phospholipids), regardless of the isotope tracer utilized. We discuss the implications of this metabolic flexibility on studies that aim to characterize apparent drug efficacy.


Subject(s)
Fatty Acids/pharmacology , Lipogenesis/drug effects , Oxygen/metabolism , Phospholipids/metabolism , Triglycerides/metabolism , Carbon Isotopes/chemistry , Cell Line, Tumor , Esterification/drug effects , Fatty Acids/chemistry , Fatty Acids/metabolism , Humans , Tandem Mass Spectrometry
5.
Am J Physiol Endocrinol Metab ; 316(6): E1105-E1117, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30912961

ABSTRACT

The regulation of nutrient homeostasis, i.e., the ability to transition between fasted and fed states, is fundamental in maintaining health. Since food is typically consumed over limited (anabolic) periods, dietary components must be processed and stored to counterbalance the catabolic stress that occurs between meals. Herein, we contrast tissue- and pathway-specific metabolic activity in fasted and fed states. We demonstrate that knowledge of biochemical kinetics that is obtained from opposite ends of the energetic spectrum can allow mechanism-based differentiation of healthy and disease phenotypes. Rat models of type 1 and type 2 diabetes serve as case studies for probing spatial and temporal patterns of metabolic activity via [2H]water labeling. Experimental designs that capture integrative whole body metabolism, including meal-induced substrate partitioning, can support an array of research surrounding metabolic disease; the relative simplicity of the approach that is discussed here should enable routine applications in preclinical models.


Subject(s)
Amino Acids/metabolism , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism , Fasting/metabolism , Fatty Acids/metabolism , Insulin/metabolism , Muscle, Skeletal/metabolism , Postprandial Period , Animals , Deuterium Oxide , Disease Models, Animal , Glycogen/metabolism , Kinetics , Lipid Metabolism/physiology , Liver/metabolism , Metabolic Networks and Pathways , Metabolomics , Rats , Rats, Wistar , Rats, Zucker , Spatio-Temporal Analysis
6.
Am J Physiol Endocrinol Metab ; 315(3): E416-E424, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29509438

ABSTRACT

Numerous studies have implicated dyslipidemia as a key factor in mediating insulin resistance. Ceramides have received special attention since their levels are inversely associated with normal insulin signaling and positively associated with factors that are involved in cardiometabolic disease. Despite the growing literature surrounding ceramide biology, there are limited data regarding the activity of ceramide synthesis and turnover in vivo. Herein, we demonstrate the ability to measure ceramide kinetics by coupling the administration of [2H]water with LC-MS/MS analyses. As a "proof-of-concept" we determined the effect of a diet-induced alteration on ceramide flux; studies also examined the effect of myriocin (a known inhibitor of serine palmitoyltransferase, the first step in sphingosine biosynthesis). Our data suggest that one can estimate ceramide synthesis and draw conclusions regarding the source of fatty acids; we discuss caveats in regards to method development in this area.


Subject(s)
Ceramides/pharmacokinetics , Animals , Chromatography, High Pressure Liquid , Deuterium Oxide/pharmacokinetics , Diet , Enzyme Inhibitors , Fatty Acids, Monounsaturated/pharmacology , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Radioactive Tracers , Serine C-Palmitoyltransferase/antagonists & inhibitors , Tandem Mass Spectrometry
8.
J Pharmacol Exp Ther ; 363(1): 80-91, 2017 10.
Article in English | MEDLINE | ID: mdl-28724692

ABSTRACT

Drug discovery and development efforts are largely based around a common expectation, namely, that direct or indirect action on a cellular process (e.g., statin-mediated enzyme inhibition or insulin-stimulated receptor activation) will have a beneficial impact on physiologic homeostasis. To expand on this, one could argue that virtually all pharmacologic interventions attempt to influence the flow of "traffic" in a biochemical network, irrespective of disease or modality. Since stable isotope tracer kinetic methods provide a measure of traffic flow (i.e., metabolic flux), their inclusion in study designs can yield novel information regarding pathway biology; the application of such methods requires the integration of knowledge in physiology, analytical chemistry, and mathematical modeling. Herein, we review the fundamental concepts that surround the use of tracer kinetics, define basic terms, and outline guiding principles via theoretical and experimental problems. Specifically, one needs to 1) recognize the types of biochemical events that change isotopic enrichments, 2) appreciate the distinction between fractional turnover and flux rate, and 3) be aware of the subtle differences between tracer kinetics and pharmacokinetics. We hope investigators can use the framework presented here to develop applications that address their specific questions surrounding biochemical flux, and thereby gain insight into the pathophysiology of disease states, and examine pharmacodynamic mechanisms.


Subject(s)
Drug Discovery/methods , Metabolic Flux Analysis/methods , Animals , Humans , Isotope Labeling , Isotopes/chemistry , Water/chemistry , Water/metabolism
9.
Am J Physiol Endocrinol Metab ; 311(6): E911-E921, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27651111

ABSTRACT

Aberrant regulation of glucose production makes a critical contribution to the impaired glycemic control that is observed in type 2 diabetes. Although isotopic tracer methods have proven to be informative in quantifying the magnitude of such alterations, it is presumed that one must rely on venous access to administer glucose tracers which therein presents obstacles for the routine application of tracer methods in rodent models. Since intraperitoneal injections are readily used to deliver glucose challenges and/or dose potential therapeutics, we hypothesized that this route could also be used to administer a glucose tracer. The ability to then reliably estimate glucose flux would require attention toward setting a schedule for collecting samples and choosing a distribution volume. For example, glucose production can be calculated by multiplying the fractional turnover rate by the pool size. We have taken a step-wise approach to examine the potential of using an intraperitoneal tracer administration in rat and mouse models. First, we compared the kinetics of [U-13C]glucose following either an intravenous or an intraperitoneal injection. Second, we tested whether the intraperitoneal method could detect a pharmacological manipulation of glucose production. Finally, we contrasted a potential application of the intraperitoneal method against the glucose-insulin clamp. We conclude that it is possible to 1) quantify glucose production using an intraperitoneal injection of tracer and 2) derive a "glucose production index" by coupling estimates of basal glucose production with measurements of fasting insulin concentration; this yields a proxy for clamp-derived assessments of insulin sensitivity of endogenous production.


Subject(s)
Blood Glucose/metabolism , Indicators and Reagents , Animals , Blood Glucose/drug effects , Carbon Isotopes , Diet, High-Fat , Female , Glucose Clamp Technique , Hypoglycemic Agents/pharmacology , Injections, Intraperitoneal , Injections, Intravenous , Insulin Resistance , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Pilot Projects , Rats , Rats, Sprague-Dawley , Rats, Zucker , Rosiglitazone , Thiazolidinediones/pharmacology
10.
Oncotarget ; 7(28): 43713-43730, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27248322

ABSTRACT

The androgen receptor (AR) plays a central role in prostate tumor growth. Inappropriate reactivation of the AR after androgen deprivation therapy promotes development of incurable castration-resistant prostate cancer (CRPC). In this study, we provide evidence that metabolic features of prostate cancer cells can be exploited to sensitize CRPC cells to AR antagonism. We identify a feedback loop between ATP-citrate lyase (ACLY)-dependent fatty acid synthesis, AMPK, and the AR in prostate cancer cells that could contribute to therapeutic resistance by maintaining AR levels. When combined with an AR antagonist, ACLY inhibition in CRPC cells promotes energetic stress and AMPK activation, resulting in further suppression of AR levels and target gene expression, inhibition of proliferation, and apoptosis. Supplying exogenous fatty acids can restore energetic homeostasis; however, this rescue does not occur through increased ß-oxidation to support mitochondrial ATP production. Instead, concurrent inhibition of ACLY and AR may drive excess ATP consumption as cells attempt to cope with endoplasmic reticulum (ER) stress, which is prevented by fatty acid supplementation. Thus, fatty acid metabolism plays a key role in coordinating ER and energetic homeostasis in CRPC cells, thereby sustaining AR action and promoting proliferation. Consistent with a role for fatty acid metabolism in sustaining AR levels in prostate cancer in vivo, AR mRNA levels in human prostate tumors correlate positively with expression of ACLY and other fatty acid synthesis genes. The ACLY-AMPK-AR network can be exploited to sensitize CRPC cells to AR antagonism, suggesting novel therapeutic opportunities for prostate cancer.


Subject(s)
ATP Citrate (pro-S)-Lyase/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Protein Kinases/metabolism , Receptors, Androgen/metabolism , AMP-Activated Protein Kinase Kinases , Androgen Receptor Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Feedback, Physiological , Humans , Male , Prostatic Neoplasms, Castration-Resistant/pathology
11.
Cancer Res ; 76(11): 3295-306, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27020861

ABSTRACT

Tamoxifen is the most widely used adjuvant chemotherapeutic for the treatment of estrogen receptor (ER)-positive breast cancer, yet a large body of clinical and preclinical data indicates that tamoxifen can modulate multiple cellular processes independently of ER status. Here, we describe the ER-independent effects of tamoxifen on tumor metabolism. Using combined pharmacologic and genetic knockout approaches, we demonstrate that tamoxifen inhibits oxygen consumption via inhibition of mitochondrial complex I, resulting in an increase in the AMP/ATP ratio and activation of the AMP-activated protein kinase (AMPK) signaling pathway in vitro and in vivo AMPK in turn promotes glycolysis and alters fatty acid metabolism. We also show that tamoxifen-induced cytotoxicity is modulated by isoform-specific effects of AMPK signaling, in which AMPKα1 promotes cell death through inhibition of the mTOR pathway and translation. By using agents that concurrently target distinct adaptive responses to tamoxifen-mediated metabolic reprogramming, we demonstrate increased cytotoxicity through synergistic therapeutic approaches. Our results demonstrate novel metabolic perturbations by tamoxifen in tumor cells, which can be exploited to expand the therapeutic potential of tamoxifen treatment beyond ER(+) breast cancer. Cancer Res; 76(11); 3295-306. ©2016 AACR.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Glycolysis/drug effects , Mitochondria, Liver/metabolism , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Female , Humans , Mice , Mice, Nude , Mitochondria, Liver/drug effects , Oxygen Consumption/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured
12.
Mol Cancer Ther ; 14(8): 1928-38, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25995437

ABSTRACT

Inhibition of the PI3K/Akt pathway decreases hypoxia within SQ20B human head and neck cancer xenografts. We set out to understand the molecular mechanism underlying this observation. We measured oxygen consumption using both a Clark electrode and an extracellular flux analyzer. We made these measurements after various pharmacologic and genetic manipulations. Pharmacologic inhibition of the PI3K/mTOR pathway or genetic inhibition of Akt/PI3K decreased the oxygen consumption rate (OCR) in vitro in SQ20B and other cell lines by 30% to 40%. Pharmacologic inhibition of this pathway increased phosphorylation of the E1α subunit of the pyruvate dehydrogenase (PDH) complex on Ser293, which inhibits activity of this critical gatekeeper of mitochondrial respiration. Expressing wild-type PTEN in a doxycycline-inducible manner in a cell line with mutant PTEN led to an increase in PDH-E1α phosphorylation and a decrease in OCR. Pretreatment of SQ20B cells with dichloroacetate (DCA), which inhibits PDH-E1α phosphorylation by inhibiting dehydrogenase kinases (PDK), reversed the decrease in OCR in response to PI3K/Akt/mTOR inhibition. Likewise, introduction of exogenous PDH-E1α that contains serine to alanine mutations, which can no longer be regulated by phosphorylation, also blunted the decrease in OCR seen with PI3K/mTOR inhibition. Our findings highlight an association between the PI3K/mTOR pathway and tumor cell oxygen consumption that is regulated in part by PDH phosphorylation. These results have important implications for understanding the effects of PI3K pathway activation in tumor metabolism and also in designing cancer therapy trials that use inhibitors of this pathway.


Subject(s)
Oxygen Consumption , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Gene Knockdown Techniques , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , TOR Serine-Threonine Kinases/antagonists & inhibitors
13.
Cancer Biol Ther ; 13(7): 575-84, 2012 May.
Article in English | MEDLINE | ID: mdl-22441776

ABSTRACT

Radiation therapy (RT) plays a critical role in the local-regional control of head and neck squamous cell carcinoma (HNSCC). However, the efficacy of RT in treating HNSCC is limited by severe normal tissue toxicity, predominantly mucositis. One pharmacological approach for increasing the clinical response to RT is the use of radiation response modifiers that preferentially sensitize tumor cells. Previously we demonstrated that curcumin, a natural plant polyphenol, increased the radiation sensitivity of HNSCC cells and that the observed sensitization was dependent on curcumin-mediated inhibition of thioredoxin reductase 1 (TxnRd1) a key cytosolic regulator of redox-dependent signaling. Here, we examined curcumin-induced radiation sensitization in HNSCC cell lines with differing HPV status and expressing different levels of TxnRd1, in vitro. The intrinsic radiation resistance of the HPV (-) cell lines was significantly higher than the HPV (+) cell lines used in our study. Notably, all of the HPV (-) cell lines expressed high levels of TxnRd1 and exhibited higher intrinsic resistance to RT. While curcumin was effective at increasing the radiation response of the resistant HPV (-) cell lines it had no effect on the HPV (+) cells. Based on these findings we employed an orthotopic, HPV (-) HNSCC tumor model in athymic nude mice to examine the effect of combining curcumin with fractionated RT, in vivo. The combination of curcumin feeding and fractionated RT had a significant effect on tumor doubling time and overall animal survival. We therefore propose that curcumin and RT should be considered as a first line treatment of HPV (-) HNSCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/virology , Curcumin/pharmacology , Head and Neck Neoplasms/virology , Papillomaviridae/genetics , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Chemoprevention , Curcumin/administration & dosage , Disease Models, Animal , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/radiotherapy , Humans , Mice , Mice, Nude , Radiation-Sensitizing Agents/administration & dosage , Thioredoxin Reductase 1/genetics , Thioredoxin Reductase 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...