Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 50
1.
PLoS One ; 18(12): e0294498, 2023.
Article En | MEDLINE | ID: mdl-38100464

BACKGROUND: Between 5-10% of patients discontinue statin therapy due to statin-associated adverse reactions, primarily statin associated muscle symptoms (SAMS). The absence of a clear clinical phenotype or of biomarkers poses a challenge for diagnosis and management of SAMS. Similarly, our incomplete understanding of the pathogenesis of SAMS hinders the identification of treatments for SAMS. Metabolomics, the profiling of metabolites in biofluids, cells and tissues is an important tool for biomarker discovery and provides important insight into the origins of symptomatology. In order to better understand the pathophysiology of this common disorder and to identify biomarkers, we undertook comprehensive metabolomic and lipidomic profiling of plasma samples from patients with SAMS who were undergoing statin rechallenge as part of their clinical care. METHODS AND FINDINGS: We report our findings in 67 patients, 28 with SAMS (cases) and 39 statin-tolerant controls. SAMS patients were studied during statin rechallenge and statin tolerant controls were studied while on statin. Plasma samples were analyzed using untargeted LC-MS metabolomics and lipidomics to detect differences between cases and controls. Differences in lipid species in plasma were observed between cases and controls. These included higher levels of linoleic acid containing phospholipids and lower ether lipids and sphingolipids. Reduced levels of acylcarnitines and altered amino acid profile (tryptophan, tyrosine, proline, arginine, and taurine) were observed in cases relative to controls. Pathway analysis identified significant increase of urea cycle metabolites and arginine and proline metabolites among cases along with downregulation of pathways mediating oxidation of branched chain fatty acids, carnitine synthesis, and transfer of acetyl groups into mitochondria. CONCLUSIONS: The plasma metabolome of patients with SAMS exhibited reduced content of long chain fatty acids and increased levels of linoleic acid (18:2) in phospholipids, altered energy production pathways (ß-oxidation, citric acid cycle and urea cycles) as well as reduced levels of carnitine, an essential mediator of mitochondrial energy production. Our findings support the hypothesis that alterations in pro-inflammatory lipids (arachidonic acid pathway) and impaired mitochondrial energy metabolism underlie the muscle symptoms of patients with statin associated muscle symptoms (SAMS).


Hydroxymethylglutaryl-CoA Reductase Inhibitors , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Prostaglandins , Muscles/metabolism , Carnitine , Fatty Acids/metabolism , Metabolomics/methods , Proline , Arginine , Biomarkers , Linoleic Acids , Urea
2.
Mol Neurobiol ; 2023 Nov 09.
Article En | MEDLINE | ID: mdl-37946007

Ischemic stroke is one of the major causes of morbidity and mortality worldwide. Mitochondria play a vital role in the pathological processes of cerebral ischemic injury, but its transplantation and underlying mechanisms remain unclear. In the present study, we examined the effects of mitochondrial therapy on the modulation of AMPK and SIRT1/PGC-1α signaling pathway, oxidative stress, and NLRP3 inflammasome activation after photothrombotic ischemic stroke (pt-MCAO). The adult male mice were subjected to the pt-MCAO in which the proximal-middle cerebral artery was exposed with a 532-nm laser beam for 4 min by retro-orbital injection of a photosensitive dye (Rose Bengal: 15 mg/kg) before the laser light exposure and isolated mitochondria (100 µg protein) were administered intranasally at 30 min, 24 h, and 48 h following post-stroke. After 72 h, mice were tested for neurobehavioral outcomes and euthanized for infarct volume, brain edema, and molecular analysis. First, we found that mitochondria therapy significantly decreased brain infarct volume and brain edema, improved neurological dysfunction, attenuated ischemic stroke-induced oxidative stress, and neuroinflammation. Second, mitochondria treatment inhibited NLRP3 inflammasome activation. Finally, mitochondria therapy accelerated p-AMPKα(Thr172) and PGC-1α expression and resorted SIRT1 protein expression levels in pt-MCAO mice. In conclusion, our results demonstrate that mitochondria therapy exerts neuroprotective effects by inhibiting oxidative damage and inflammation, mainly dependent on the heightening activation of the AMPK and SIRT1/PGC-1α signaling pathway. Thus, intranasal delivery of mitochondria might be considered a new therapeutic strategy for ischemic stroke treatment.

3.
Adv Exp Med Biol ; 1438: 33-36, 2023.
Article En | MEDLINE | ID: mdl-37845436

Hypoxia-inducible factor 1 (HIF-1) is a major player in the oxygen sensor system as well as a transcription factor. HIF-1 is also associated in the pathogenesis of many brain diseases including Alzheimer's disease (AD), epilepsy and stroke. HIF-1 regulates the expression of many genes such as those involved in glycolysis, erythropoiesis, angiogenesis and proliferation in hypoxic condition. Despite several studies, the mechanism through which HIF-1 confers neuroprotection remains unclear, one of them is modulating metabolic profiles and inflammatory pathways. Characterization of the neuroprotective role of HIF-1 may be through its stabilization and the regulation of target genes that aid in the early adaptation to the oxidative stressors. It is interesting to note that mounting data from recent years point to an additional crucial regulatory role for hypoxia-inducible factors (HIFs) in inflammation. HIFs in immune cells regulate the production of glycolytic energy as well as innate immunity, pro-inflammatory gene expression, and mediates activation of pro-survival pathways. The present review highlights the contribution of HIF-1 to neuroprotection where inflammation is the crucial factor in the pathogenesis contributing to neural death. The potential mechanisms that contribute to neuroprotection as a result of the downstream targets of HIF-1α are discussed. Such mechanisms include those mediated through IL-10, an anti-inflammatory molecule involved in activating pro-survival signaling mechanisms via AKT/ERK and JAK/STAT pathways.


Gene Expression Regulation , Neuroprotection , Humans , Signal Transduction , Inflammation/genetics , Phenotype , Hypoxia-Inducible Factor 1, alpha Subunit/genetics
4.
Metabolism ; 144: 155589, 2023 07.
Article En | MEDLINE | ID: mdl-37182789

BACKGROUND: Evidence is accumulating that growth hormone (GH) protects against the development of steatosis and progression of non-alcoholic fatty liver disease (NAFLD). GH may control steatosis indirectly by altering systemic insulin sensitivity and substrate delivery to the liver and/or by the direct actions of GH on hepatocyte function. APPROACH: To better define the hepatocyte-specific role of GH receptor (GHR) signaling on regulating steatosis, we used a mouse model with adult-onset, hepatocyte-specific GHR knockdown (aHepGHRkd). To prevent the reduction in circulating insulin-like growth factor 1 (IGF1) and the subsequent increase in GH observed after aHepGHRkd, subsets of aHepGHRkd mice were treated with adeno-associated viral vectors (AAV) driving hepatocyte-specific expression of IGF1 or a constitutively active form of STAT5b (STAT5bCA). The impact of hepatocyte-specific modulation of GHR, IGF1 and STAT5b on carbohydrate and lipid metabolism was studied across multiple nutritional states and in the context of hyperinsulinemic:euglycemic clamps. RESULTS: Chow-fed male aHepGHRkd mice developed steatosis associated with an increase in hepatic glucokinase (GCK) and ketohexokinase (KHK) expression and de novo lipogenesis (DNL) rate, in the post-absorptive state and in response to refeeding after an overnight fast. The aHepGHRkd-associated increase in hepatic KHK, but not GCK and steatosis, was dependent on hepatocyte expression of carbohydrate response element binding protein (ChREBP), in re-fed mice. Interestingly, under clamp conditions, aHepGHRkd also increased the rate of DNL and expression of GCK and KHK, but impaired insulin-mediated suppression of hepatic glucose production, without altering plasma NEFA levels. These effects were normalized with AAV-mediated hepatocyte expression of IGF1 or STAT5bCA. Comparison of the impact of AAV-mediated hepatocyte IGF1 versus STAT5bCA in aHepGHRkd mice across multiple nutritional states, indicated the restorative actions of IGF1 are indirect, by improving systemic insulin sensitivity, independent of changes in the liver transcriptome. In contrast, the actions of STAT5b are due to the combined effects of raising IGF1 and direct alterations in the hepatocyte gene program that may involve suppression of BCL6 and FOXO1 activity. However, the direct and IGF1-dependent actions of STAT5b cannot fully account for enhanced GCK activity and lipogenic gene expression observed after aHepGHRkd, suggesting other GHR-mediated signals are involved. CONCLUSION: These studies demonstrate hepatocyte GHR-signaling controls hepatic glycolysis, DNL, steatosis and hepatic insulin sensitivity indirectly (via IGF1) and directly (via STAT5b). The relative contribution of these indirect and direct actions of GH on hepatocytes is modified by insulin and nutrient availability. These results improve our understanding of the physiologic actions of GH on regulating adult metabolism to protect against NAFLD progression.


Human Growth Hormone , Insulin Resistance , Non-alcoholic Fatty Liver Disease , Male , Mice , Animals , Lipogenesis/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , Insulin Resistance/physiology , Liver/metabolism , Growth Hormone/metabolism , Insulin/metabolism , Glycolysis , Glucose/metabolism , Human Growth Hormone/metabolism
5.
Adv Exp Med Biol ; 1395: 75-79, 2022.
Article En | MEDLINE | ID: mdl-36527617

Hypoxia inducible factor alpha (HIF1α) is associated with neuroprotection conferred by diet-induced ketosis but the underlying mechanism remains unclear. In this study we use a ketogenic diet in rodents to induce a metabolic state of chronic ketosis, as measured by elevated blood ketone bodies. Chronic ketosis correlates with neuroprotection in both aged and following focal cerebral ischaemia and reperfusion (via middle cerebral artery occlusion, MCAO) in mouse and rat models. Ketone bodies are known to be used efficiently by the brain and metabolism of ketone bodies is associated with increased cytosolic succinate levels that inhibits prolyl hydroxylases allowing HIF1α to accumulate. Ketosis also regulates inflammatory pathways, and HIF1α is reported to be essential for gene expression of interleukin10 (IL10). Therefore we hypothesised that ketosis-stabilised HIF1α modulates the expression of inflammatory cytokines orchestrating neuroprotection. To test changes in cytokine levels in rodent brain, eight-week-old rats were fed either the standard chow diet (SD) or the ketogenic (KG) diet for 4 weeks before ischaemia experiments (MCAO) were performed and the brain tissues were collected. Consistent with our hypothesis, immunoblotting analysis shows IL10 levels were significantly higher in KG diet rat brain compared to SD, whereas the TNFα and IL6 levels were significantly lower in the brains of KG diet fed group.


Diet, Ketogenic , Ketosis , Animals , Rats , Mice , Interleukin-10/genetics , Interleukin-10/metabolism , Ketosis/metabolism , Ketone Bodies/metabolism , Brain/metabolism
6.
Nat Commun ; 13(1): 6062, 2022 10 13.
Article En | MEDLINE | ID: mdl-36229459

Almost all effective treatments for non-alcoholic fatty liver disease (NAFLD) involve reduction of adiposity, which suggests the metabolic axis between liver and adipose tissue is essential to NAFLD development. Since excessive dietary sugar intake may be an initiating factor for NAFLD, we have characterized the metabolic effects of liquid sucrose intake at concentrations relevant to typical human consumption in mice. We report that sucrose intake induces sexually dimorphic effects in liver, adipose tissue, and the microbiome; differences concordant with steatosis severity. We show that when steatosis is decoupled from impairments in insulin responsiveness, sex is a moderating factor that influences sucrose-driven lipid storage and the contribution of de novo fatty acid synthesis to the overall hepatic triglyceride pool. Our findings provide physiologic insight into how sex influences the regulation of adipose-liver crosstalk and highlight the importance of extrahepatic metabolism in the pathogenesis of diet-induced steatosis and NAFLD.


Non-alcoholic Fatty Liver Disease , Adipose Tissue/metabolism , Animals , Dietary Sucrose/adverse effects , Fatty Acids/metabolism , Humans , Insulin/metabolism , Lipid Metabolism , Liver/metabolism , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Triglycerides/metabolism
7.
J Stroke Cerebrovasc Dis ; 31(2): 106226, 2022 Feb.
Article En | MEDLINE | ID: mdl-34847489

OBJECTIVES: Acute hyperglycemia (HG) exacerbates reperfusion injury after stroke. Our recent studies showed that acute HG upregulates thioredoxin-interacting protein (TXNIP) expression, which in turn induces inflammation and neurovascular damage in a suture model of ischemic stroke. The aim of the present study was to investigate the effect of acute HG on TXNIP-associated neurovascular damage, in a more clinically relevant murine model of embolic stroke and intravenous tissue plasminogen activator (IV-tPA) reperfusion. MATERIALS AND METHODS: HG was induced in adult male mice, by intraperitoneal injection of 20% glucose. This was followed by embolic middle cerebral artery occlusion (eMCAO), with or without IV-tPA (10 mg/kg) given 3 h post embolization. Brain infarction, edema, hemoglobin content, expression of matrix metalloproteinase (MMP-9), vascular endothelial growth factor A (VEGFA), tight junction proteins (claudin-5, occluding, and zonula occludens-1), TXNIP, and NOD-like receptor protein3 (NLRP3)-inflammasome activation were evaluated at 24 h after eMCAO. RESULTS: HG alone significantly increased TXNIP in the brain after eMCAO, and this was associated with exacerbated hemorrhagic transformation (HT; as measured by hemoglobin content). IV-tPA in HG conditions showed a trend to decrease infarct volume, but worsened HT after eMCAO, suggesting that HG reduces the therapeutic efficacy of IV-tPA. Further, HG and tPA-reperfusion did not show significant differences in expression of MMP-9, VEGFA, junction proteins, and NLRP3 inflammasome activation between the groups. CONCLUSION: The current findings suggest a potential role for TXNIP in the occurrence of HT in hyperglycemic conditions following eMCAO. Further studies are needed to understand the precise role of vascular TXNIP on HG/tPA-induced neurovascular damage after stroke.


Embolic Stroke , Hyperglycemia , Reperfusion , Tissue Plasminogen Activator , Animals , Carrier Proteins/physiology , Disease Models, Animal , Embolic Stroke/drug therapy , Embolic Stroke/pathology , Hyperglycemia/complications , Inflammasomes/physiology , Injections, Intravenous , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Thioredoxins/physiology , Tissue Plasminogen Activator/administration & dosage
8.
Int J Mol Sci ; 22(24)2021 Dec 16.
Article En | MEDLINE | ID: mdl-34948286

Di(2-ethylhexyl) phthalate (DEHP) is commonly used as a plasticizer in various industrial and household plastic products, ensuring widespread human exposures. Its routine detection in human bio-fluids and the propensity of its monoester metabolite to activate peroxisome proliferator activated receptor-α (PPARα) and perturb lipid metabolism implicate it as a metabolic disrupter. In this study we evaluated the effects of DEHP exposure on hepatic levels of free CoA and various CoA esters, while also confirming the metabolic activation to CoA esters and partial ß-oxidation of a DEHP metabolite (2-ethyhexanol). Male Wistar rats were exposed via diet to 2% (w/w) DEHP for fourteen-days, following which hepatic levels of free CoA and various CoA esters were identified using liquid chromatography-mass spectrometry. DEHP exposed rats showed significantly elevated free CoA and increased levels of physiological, DEHP-derived and unidentified CoA esters. The physiological CoA ester of malonyl-CoA and DEHP-derived CoA ester of 3-keto-2-ethylhexanoyl-CoA were the most highly elevated, at eighteen- and ninety eight-times respectively. We also detected sixteen unidentified CoA esters which may be derivative of DEHP metabolism or induction of other intermediary metabolism metabolites. Our results demonstrate that DEHP is a metabolic disrupter which affects production and sequestration of CoA, an essential cofactor of oxidative and biosynthetic reactions.


Coenzyme A/metabolism , Diethylhexyl Phthalate/metabolism , Liver/metabolism , Phthalic Acids/metabolism , Animals , Lipid Metabolism/physiology , Male , Oxidation-Reduction , PPAR alpha/metabolism , Plasticizers/metabolism , Rats , Rats, Wistar
9.
FASEB J ; 35(12): e22018, 2021 12.
Article En | MEDLINE | ID: mdl-34731499

Adipose tissue is the primary site of energy storage, playing important roles in health. While adipose research largely focuses on obesity, fat also has other critical functions, producing adipocytokines and contributing to normal nutrient metabolism, which in turn play important roles in satiety and total energy homeostasis. SMAD2/3 proteins are downstream mediators of activin signaling, which regulate critical preadipocyte and mature adipocyte functions. Smad2 global knockout mice exhibit embryonic lethality, whereas global loss of Smad3 protects mice against diet-induced obesity. The direct contributions of Smad2 and Smad3 in adipose tissues, however, are unknown. Here, we sought to determine the primary effects of adipocyte-selective reduction of Smad2 or Smad3 on diet-induced adiposity using Smad2 or Smad3 "floxed" mice intercrossed with Adiponectin-Cre mice. Additionally, we examined visceral and subcutaneous preadipocyte differentiation efficiency in vitro. Almost all wild type subcutaneous preadipocytes differentiated into mature adipocytes. In contrast, visceral preadipocytes differentiated poorly. Exogenous activin A suppressed differentiation of preadipocytes from both depots. Smad2 conditional knockout (Smad2cKO) mice did not exhibit significant effects on weight gain, irrespective of diet, whereas Smad3 conditional knockout (Smad3cKO) male mice displayed a trend of reduced body weight on high-fat diet. On both diets, Smad3cKO mice displayed an adipose depot-selective phenotype, with a significant reduction in subcutaneous fat mass but not visceral fat mass. Our data suggest that Smad3 is an important contributor to the maintenance of subcutaneous white adipose tissue in a sex-selective fashion. These findings have implications for understanding SMAD-mediated, depot selective regulation of adipocyte growth and differentiation.


Adipogenesis , Adipose Tissue, White/cytology , Adiposity , Intra-Abdominal Fat/cytology , Smad2 Protein/physiology , Smad3 Protein/physiology , Subcutaneous Fat/cytology , Activins/genetics , Activins/metabolism , Adipose Tissue, White/metabolism , Animals , Cell Differentiation , Diet, High-Fat , Female , Intra-Abdominal Fat/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Subcutaneous Fat/metabolism
10.
Redox Biol ; 47: 102163, 2021 11.
Article En | MEDLINE | ID: mdl-34655995

12/15-lipoxygenase (12/15-LOX) plays an essential role in oxidative conversion of polyunsaturated fatty acids into various bioactive lipid molecules. Although 12/15-LOX's role in the pathophysiology of various human diseases has been well studied, its role in weight gain is controversial and poorly clarified. Here, we demonstrated the role of 12/15-LOX in high-fat diet (HFD)-induced weight gain in a mouse model. We found that 12/15-LOX mediates HFD-induced de novo lipogenesis (DNL), triglyceride (TG) biosynthesis and the transport of TGs from the liver to adipose tissue leading to white adipose tissue (WAT) expansion and weight gain via xanthine oxidase (XO)-dependent production of H2O2. 12/15-LOX deficiency leads to cullin2-mediated ubiquitination and degradation of XO, thereby suppressing H2O2 production, DNL and TG biosynthesis resulting in reduced WAT expansion and weight gain. These findings infer that manipulation of 12/15-LOX metabolism may manifest a potential therapeutic target for weight gain and obesity.


Lipogenesis , Xanthine Oxidase , Animals , Arachidonate 15-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/metabolism , Diet, High-Fat/adverse effects , Hydrogen Peroxide/metabolism , Liver/metabolism , Mice , Triglycerides/metabolism , Weight Gain , Xanthine Oxidase/metabolism
11.
Pharmaceuticals (Basel) ; 14(10)2021 Sep 27.
Article En | MEDLINE | ID: mdl-34681207

We hypothesize that endothelial-specific thioredoxin-interacting protein knock-out (EC-TXNIP KO) mice will be more resistant to the neurovascular damage (hemorrhagic-transformation-HT) associated with hyperglycemia (HG) in embolic stroke. Adult-male EC-TXNIP KO and wild-type (WT) littermate mice were injected with-streptozotocin (40 mg/kg, i.p.) for five consecutive days to induce diabetes. Four-weeks after confirming HG, mice were subjected to embolic middle cerebral artery occlusion (eMCAO) followed by tissue plasminogen activator (tPA)-reperfusion (10 mg/kg at 3 h post-eMCAO). After the neurological assessment, animals were sacrificed at 24 h for neurovascular stroke outcomes. There were no differences in cerebrovascular anatomy between the strains. Infarct size, edema, and HT as indicated by hemoglobin (Hb)-the content was significantly higher in HG-WT mice, with or without tPA-reperfusion, compared to normoglycemic WT mice. Hyperglycemic EC-TXNIP KO mice treated with tPA tended to show lower Hb-content, edema, infarct area, and less hemorrhagic score compared to WT hyperglycemic mice. EC-TXNIP KO mice showed decreased expression of inflammatory mediators, apoptosis-associated proteins, and nitrotyrosine levels. Further, vascular endothelial growth factor-A and matrix-metalloproteinases (MMP-9/MMP-3), which degrade junction proteins and increase blood-brain-barrier permeability, were decreased in EC-TXNIP KO mice. Together, these findings suggest that vascular-TXNIP could be a novel therapeutic target for neurovascular damage after stroke.

12.
Adv Exp Med Biol ; 1269: 3-7, 2021.
Article En | MEDLINE | ID: mdl-33966187

Hypoxia inducible factor alpha (HIF1α) is associated with neuroprotection conferred by diet-induced ketosis, but the underlying mechanism remains unclear. In this study, we use a ketogenic diet in rodents to induce a metabolic state of chronic ketosis, as measured by elevated blood ketone bodies. Chronic ketosis correlates with neuroprotection in both aged and following focal cerebral ischemia and reperfusion (via middle cerebral artery occlusion, MCAO) in mouse and rat models. Ketone bodies are known to be used efficiently by the brain, and metabolism of ketone bodies is associated with increased cytosolic succinate levels that inhibits prolyl hydroxylases allowing HIF1α to accumulate. Ketosis also regulates inflammatory pathways, and HIF1α is reported to be essential for gene expression of interleukin 10 (IL10). Therefore, we hypothesized that ketosis-stabilized HIF1α modulates the expression of inflammatory cytokines orchestrating neuroprotection. To test changes in cytokine levels in rodent brain, 8-week-rats were fed either the standard chow diet (SD) or the KG diet for 4 weeks before ischemia experiments (MCAO) were performed and the brain tissues were collected. Consistent with our hypothesis, immunoblotting analysis shows IL10 levels were significantly higher in KG diet rat brain compared to SD, whereas the TNFα and IL6 levels were significantly lower in the brains of KG diet-fed group.


Brain Ischemia , Diet, Ketogenic , Ketosis , Animals , Brain , Ketone Bodies , Mice , Rats
13.
J Immunol ; 202(4): 1265-1286, 2019 02 15.
Article En | MEDLINE | ID: mdl-30659108

Macrophages (MΦs) are heterogeneous and metabolically flexible, with metabolism strongly affecting immune activation. A classic response to proinflammatory activation is increased flux through glycolysis with a downregulation of oxidative metabolism, whereas alternative activation is primarily oxidative, which begs the question of whether targeting glucose metabolism is a viable approach to control MΦ activation. We created a murine model of myeloid-specific glucose transporter GLUT1 (Slc2a1) deletion. Bone marrow-derived MΦs (BMDM) from Slc2a1M-/- mice failed to uptake glucose and demonstrated reduced glycolysis and pentose phosphate pathway activity. Activated BMDMs displayed elevated metabolism of oleate and glutamine, yet maximal respiratory capacity was blunted in MΦ lacking GLUT1, demonstrating an incomplete metabolic reprogramming. Slc2a1M-/- BMDMs displayed a mixed inflammatory phenotype with reductions of the classically activated pro- and anti-inflammatory markers, yet less oxidative stress. Slc2a1M-/- BMDMs had reduced proinflammatory metabolites, whereas metabolites indicative of alternative activation-such as ornithine and polyamines-were greatly elevated in the absence of GLUT1. Adipose tissue MΦs of lean Slc2a1M-/- mice had increased alternative M2-like activation marker mannose receptor CD206, yet lack of GLUT1 was not a critical mediator in the development of obesity-associated metabolic dysregulation. However, Ldlr-/- mice lacking myeloid GLUT1 developed unstable atherosclerotic lesions. Defective phagocytic capacity in Slc2a1M-/- BMDMs may have contributed to unstable atheroma formation. Together, our findings suggest that although lack of GLUT1 blunted glycolysis and the pentose phosphate pathway, MΦ were metabolically flexible enough that inflammatory cytokine release was not dramatically regulated, yet phagocytic defects hindered MΦ function in chronic diseases.


Disease Models, Animal , Glucose Transporter Type 1/metabolism , Macrophages/metabolism , Animals , Glucose Transporter Type 1/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype
14.
Am J Physiol Endocrinol Metab ; 315(6): E1168-E1184, 2018 12 01.
Article En | MEDLINE | ID: mdl-30253111

Brain-derived neurotrophic factor (BDNF) is a key neuropeptide in the central regulation of energy balance. The Bdnf gene contains nine promoters, each producing specific mRNA transcripts that encode a common protein. We sought to assess the phenotypic outcomes of disrupting BDNF production from individual Bdnf promoters. Mice with an intact coding region but selective disruption of BDNF production from Bdnf promoters I, II, IV, or VI (Bdnf-e1-/-, -e2-/-, -e4-/-, and -e6-/-) were created by inserting an enhanced green fluorescent protein-STOP cassette upstream of the targeted promoter splice donor site. Body composition was measured by MRI weekly from age 4 to 22 wk. Energy expenditure was measured by indirect calorimetry at 18 wk. Food intake was measured in Bdnf-e1-/- and Bdnf-e2-/- mice, and pair feeding was conducted. Weight gain, lean mass, fat mass, and percent fat of Bdnf-e1-/- and Bdnf-e2-/- mice (both sexes) were significantly increased compared with wild-type littermates. For Bdnf-e4-/- and Bdnf-e6-/- mice, obesity was not observed with either chow or high-fat diet. Food intake was increased in Bdnf-e1-/- and Bdnf-e2-/- mice, and pair feeding prevented obesity. Mutant and wild-type littermates for each strain (both sexes) had similar total energy expenditure after adjustment for body composition. These findings suggest that the obesity phenotype observed in Bdnf-e1-/- and Bdnf-e2-/- mice is attributable to hyperphagia and not altered energy expenditure. Our findings show that disruption of BDNF from specific promoters leads to distinct body composition effects, with disruption from promoters I or II, but not IV or VI, inducing obesity.


Body Composition/genetics , Body Weight/genetics , Brain-Derived Neurotrophic Factor/genetics , Obesity/genetics , Promoter Regions, Genetic , Animals , Brain-Derived Neurotrophic Factor/metabolism , Calorimetry, Indirect , Eating/genetics , Energy Metabolism/genetics , Mice , Mice, Transgenic , Obesity/metabolism , Phenotype
15.
Adv Exp Med Biol ; 1072: 77-82, 2018.
Article En | MEDLINE | ID: mdl-30178327

Transient global brain ischemia, induced by cardiac arrest and resuscitation, results in reperfusion injury leading to delayed selective neuronal cell loss and post-resuscitation mortality. This study determined the effects of post-resuscitation hypotension and hypothermia on long-term survival following cardiac arrest and resuscitation. The capillary density was also determined. Based on the mean arterial blood pressure (MABP) at 1 h of recovery, the normotension group (MABP 80-120 mmHg) and hypotension group (MABP <80 mmHg) were defined. The overall survival was determined at 4 days of recovery. Brain microvascular density was assessed using immunohistochemistry of the glucose transporter, GLUT-1. The pre-arrest MABP was similar in each group; at 1 h after resuscitation, the MABP in the normotension groups was about 80% of their pre-arrest values; the hypotension group had a significantly lower MABP compared to the normotension group. The overall survival rate was lower in the hypotension group compared to the normotension group (36%, 4/11 vs. 67%, 14/21) under the normothermic condition. Brain blood flow in the hypotension group was lower (33% decrease) compared to the normotension group at 1-h post-resuscitation. Compared to the pre-arrest baseline, the capillary density was significantly increased at 14 days of recovery (355 ± 42 vs. 469 ± 50, number/mm2) in the cortex. The capillary density in hippocampus was also increased at 4-30 days following cardiac arrest and resuscitation. Our results suggest that rats able to maintain their post-resuscitation blood pressure at normotension, had higher brain blood flow during the early recovery phase, and improved survival outcome following cardiac arrest and resuscitation. In addition, cardiac arrest and resuscitation induced angiogenesis in brain in the first month of recovery.


Brain Ischemia/etiology , Brain/blood supply , Heart Arrest/complications , Hypotension/etiology , Animals , Arterial Pressure/physiology , Brain Ischemia/pathology , Cardiopulmonary Resuscitation , Heart Arrest/physiopathology , Hypotension/physiopathology , Male , Rats , Rats, Inbred F344 , Reperfusion Injury/etiology , Reperfusion Injury/pathology
16.
J Biol Chem ; 293(9): 3399-3409, 2018 03 02.
Article En | MEDLINE | ID: mdl-29317502

Phosphoenolpyruvate carboxykinase (Pck1) is a metabolic enzyme that is integral to the gluconeogenic and glyceroneogenic pathways. However, Pck1's role in macrophage metabolism and function is unknown. Using stable isotopomer MS analysis in a mouse model with a myeloid cell-specific Pck1 deletion, we show here that this deletion increases the proinflammatory phenotype in macrophages. Incubation of LPS-stimulated bone marrow-derived macrophages (BMDM) with [U-13C]glucose revealed reduced 13C labeling of citrate and malate and increased 13C labeling of lactate in Pck1-deleted bone marrow-derived macrophages. We also found that the Pck1 deletion in the myeloid cells increases reactive oxygen species (ROS). Of note, this altered macrophage metabolism increased expression of the M1 cytokines TNFα, IL-1ß, and IL-6. We therefore conclude that Pck1 contributes to M1 polarization in macrophages. Our findings provide important insights into the factors determining the macrophage inflammatory response and indicate that Pck1 activity contributes to metabolic reprogramming and polarization in macrophages.


Gene Deletion , Macrophages/enzymology , Phenotype , Phosphoenolpyruvate Carboxykinase (GTP)/deficiency , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Animals , Cell Polarity , Glucose/metabolism , Glutamine/metabolism , Inflammation/enzymology , Inflammation/genetics , Inflammation/immunology , Macrophages/cytology , Macrophages/metabolism , Mice , Palmitic Acid/metabolism , RAW 264.7 Cells
17.
Adv Exp Med Biol ; 977: 205-213, 2017.
Article En | MEDLINE | ID: mdl-28685447

Over the past decade we have consistently shown that ketosis is neuroprotective against ischemic insults in rats. We reported that diet-induced ketotic rats had a significant reduction in infarct volume when subjected to middle cerebral artery occlusion (MCAO), and improved survival and recovery after cardiac arrest and resuscitation. The neuroprotective mechanisms of ketosis (via ketogenic diet; KG) include (i) ketones are alternate energy substrates that can restore energy balance when glucose metabolism is deficient and (ii) ketones modulate cell-signalling pathways that are cytoprotective. We investigated the effects of diet-induced ketosis following transient focal cerebral ischemia in mice. The correlation between levels of ketosis and hypoxic inducible factor-1alpha (HIF-1α), AKT (also known as protein kinase B or PKB) and 5' AMP-activated protein kinase (AMPK) were determined. Mice were fed with KG diet or standard lab-chow (STD) diet for 4 weeks. For the MCAO group, mice underwent 60 min of MCAO and total brain infarct volumes were evaluated 48 h after reperfusion. In a separate group of mice, brain tissue metabolites, levels of HIF-1α, phosphorylated AKT (pAKT), and AMPK were measured. After feeding a KG diet, levels of blood ketone bodies (beta-hydroxyburyrate, BHB) were increased. There was a proportional decrease in infarct volumes with increased blood BHB levels (KG vs STD; 4.2 ± 0.6 vs 7.8 ± 2.2 mm3, mean ± SEM). A positive correlation was also observed with HIF-1α and pAKT relative to blood BHB levels. Our results showed that chronic ketosis can be induced in mice by KG diet and was neuroprotective against focal cerebral ischemia in a concentration dependent manner. Potential mechanisms include upregulation of cytoprotective pathways such as those associated with HIF-1α, pAKT and AMPK.


Brain Ischemia/prevention & control , Diet, Ketogenic , Infarction, Middle Cerebral Artery/diet therapy , Ketosis/pathology , Animals , Brain Ischemia/etiology , Disease Models, Animal , Feeding Behavior/physiology , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Ischemic Attack, Transient/etiology , Ischemic Attack, Transient/prevention & control , Ketosis/etiology , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents
18.
J Neurochem ; 141(2): 162-164, 2017 04.
Article En | MEDLINE | ID: mdl-28299805

Read the highlighted article 'Effects of a dietary ketone ester on hippocampal glycolytic and tricarboxylic acid cycle intermediates and amino acids in a 3xTgAD mouse model of Alzheimer's disease' on page 195.


Alzheimer Disease , Citric Acid Cycle , Amino Acids , Animals , Hippocampus , Humans , Mice , Mice, Transgenic
19.
Adv Exp Med Biol ; 923: 31-36, 2016.
Article En | MEDLINE | ID: mdl-27526121

In this study we investigated the effect of Dl-3-n-butylphthalide (NBP), a clinically used drug for stroke patients in China, on the recovery following cardiac arrest and resuscitation in rats. Male Wistar rats (3-month old) underwent cardiac arrest (12 min) and resuscitation. Rats were randomly assigned to the following groups: sham non-arrested group, vehicle group (vehicle-treated, 7 days before cardiac arrest and 4 days post-resuscitation), NBP pre-treated group (NBP-treated, 7 days before cardiac arrest), and NBP post-treated group (NBP-treated, 4 days post-resuscitation). Overall survival rates and hippocampal neuronal counts were determined in each group at 4 days post-resuscitation. Results showed that NBP pre-treated group (80 %) and NBP post-treated group (86 %) had significantly higher survival rates compared to that of the vehicle group (50 %). At 4 days of recovery, only about 20 % of hippocampal neurons were preserved in the vehicle group compared to the sham non-arrested group. The hippocampal CA1 cell counts in the NBP pre-treated group and NBP post-treated group were significantly higher than the counts in the vehicle group, about 50-60 % of the counts of non-arrested rats. The data suggest that NBP has both preventive and therapeutic effect on improving outcome following cardiac arrest and resuscitation, and NBP might be a potential early phase treatment for patients recovered from cardiac arrest and resuscitation.


Benzofurans/pharmacology , CA1 Region, Hippocampal/drug effects , Heart Arrest/therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Resuscitation , Animals , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiopathology , Disease Models, Animal , Heart Arrest/pathology , Heart Arrest/physiopathology , Male , Neurons/pathology , Rats, Wistar , Recovery of Function , Time Factors
20.
Adv Exp Med Biol ; 876: 265-270, 2016.
Article En | MEDLINE | ID: mdl-26782221

In this study we investigated the effect of aging on brain blood flow following transient global ischemia. Male Fisher rats (6 and 24 months old) underwent cardiac arrest (15 min) and resuscitation. Regional brain (cortex, hippocampus, brainstem and cerebellum) blood flow was measured in non-arrested rats and 1-h recovery rats using [14C] iodoantipyrene (IAP) autoradiography; the 4-day survival rate was determined in the two age groups. The pre-arrest baseline blood flows were similar in cortex, brainstem and cerebellum between the 6-month and the 24-month old rats; however, the baseline blood flow in hippocampus was significantly lower in the 24-month old group. At 1 h following cardiac arrest and resuscitation, both 6-month and 24-month groups had significantly lower blood flows in all regions than the pre-arrest baseline values; compared to the 6-month old group, the blood flow was significantly lower (about 40% lower) in all regions in the 24-month old group. The 4-day survival rate for the 6-month old rats was 50% (3/6) whereas none of the 24-month old rats (0/10) survived for 4 days. The data suggest that there is an increased vulnerability to brain ischemic-reperfusion injury in the aged rats; the degree of post-recovery hypoperfusion may contribute to the high mortality in the aged rats following cardiac arrest and resuscitation.


Aging/physiology , Cardiopulmonary Resuscitation , Heart Arrest/mortality , Animals , Cerebrovascular Circulation , Heart Arrest/physiopathology , Male , Rats , Rats, Inbred F344 , Reperfusion Injury/etiology
...