57 results on '"Puchowicz MA"'
Search Results
2. Intranasal Delivery of Mitochondria Attenuates Brain Injury by AMPK and SIRT1/PGC-1α Pathways in a Murine Model of Photothrombotic Stroke.
- Author
-
Salman M, Stayton AS, Parveen K, Parveen A, Puchowicz MA, Parvez S, Bajwa A, and Ishrat T
- Subjects
- Animals, Male, Mice, Oxidative Stress drug effects, Mice, Inbred C57BL, Brain Injuries pathology, Brain Injuries drug therapy, Brain Injuries metabolism, Inflammasomes metabolism, Infarction, Middle Cerebral Artery complications, Infarction, Middle Cerebral Artery pathology, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Thrombosis pathology, Sirtuin 1 metabolism, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha metabolism, Administration, Intranasal, Mitochondria metabolism, Mitochondria drug effects, Disease Models, Animal, Signal Transduction drug effects, AMP-Activated Protein Kinases metabolism, Stroke pathology, Stroke drug therapy, Stroke metabolism
- Abstract
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., (© 2023. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
3. Effect of statin treatment on metabolites, lipids and prostanoids in patients with Statin Associated Muscle Symptoms (SAMS).
- Author
-
Garrett TJ, Puchowicz MA, Park EA, Dong Q, Farage G, Childress R, Guingab J, Simpson CL, Sen S, Brogdon EC, Buchanan LM, Raghow R, and Elam MB
- Subjects
- Humans, Prostaglandins, Muscles metabolism, Carnitine, Fatty Acids metabolism, Metabolomics methods, Proline, Arginine, Biomarkers, Linoleic Acids, Urea, Hydroxymethylglutaryl-CoA Reductase Inhibitors adverse effects
- Abstract
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 (β-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)., Competing Interests: The authors have declared that no competing interests exist., (Copyright: This is an open access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication.)
- Published
- 2023
- Full Text
- View/download PDF
4. Direct and systemic actions of growth hormone receptor (GHR)-signaling on hepatic glycolysis, de novo lipogenesis and insulin sensitivity, associated with steatosis.
- Author
-
Vázquez-Borrego MC, Del Río-Moreno M, Pyatkov M, Sarmento-Cabral A, Mahmood M, Pelke N, Wnek M, Cordoba-Chacon J, Waxman DJ, Puchowicz MA, McGuinness OP, and Kineman RD
- Subjects
- Male, Mice, Animals, Lipogenesis genetics, Receptors, Somatotropin genetics, Receptors, Somatotropin metabolism, Liver metabolism, Growth Hormone metabolism, Insulin metabolism, Glycolysis, Glucose metabolism, Non-alcoholic Fatty Liver Disease metabolism, Insulin Resistance physiology, Human Growth Hormone metabolism
- Abstract
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 (STAT5b
CA ). 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., Competing Interests: Declaration of competing interest Authors have nothing to disclose., (Copyright © 2023. Published by Elsevier Inc.)- Published
- 2023
- Full Text
- View/download PDF
5. Pro-survival Phenotype of HIF-1α: Neuroprotection Through Inflammatory Mechanisms.
- Author
-
Puchowicz MA, Parveen K, Sethuraman A, Ishrat T, Xu K, and LaManna J
- Subjects
- Humans, Signal Transduction, Inflammation genetics, Phenotype, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Neuroprotection, Gene Expression Regulation
- Abstract
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., (© 2023. The Author(s), under exclusive license to Springer Nature Switzerland AG.)
- Published
- 2023
- Full Text
- View/download PDF
6. Chronic intake of high dietary sucrose induces sexually dimorphic metabolic adaptations in mouse liver and adipose tissue.
- Author
-
Stephenson EJ, Stayton AS, Sethuraman A, Rao PK, Meyer A, Gomes CK, Mulcahy MC, McAllan L, Puchowicz MA, Pierre JF, Bridges D, and Han JC
- Subjects
- Adipose Tissue metabolism, Animals, Dietary Sucrose adverse effects, Fatty Acids metabolism, Humans, Insulin metabolism, Lipid Metabolism, Liver metabolism, Mice, Triglycerides metabolism, Non-alcoholic Fatty Liver Disease metabolism
- Abstract
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., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
7. Acute Hyperglycemia Exacerbates Hemorrhagic Transformation after Embolic Stroke and Reperfusion with tPA: A Possible Role of TXNIP-NLRP3 Inflammasome.
- Author
-
Salman M, Ismael S, Li L, Ahmed HA, Puchowicz MA, and Ishrat T
- Subjects
- Animals, Carrier Proteins physiology, Disease Models, Animal, Inflammasomes physiology, Injections, Intravenous, Male, Mice, NLR Family, Pyrin Domain-Containing 3 Protein physiology, Thioredoxins physiology, Embolic Stroke drug therapy, Embolic Stroke pathology, Hyperglycemia complications, Reperfusion, Tissue Plasminogen Activator administration & dosage
- Abstract
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., Competing Interests: Conflicts of Interest The authors declare no conflict of interest., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
8. Chronic Ketosis Modulates HIF1α-Mediated Inflammatory Response in Rat Brain.
- Author
-
Sethuraman A, Rao P, Pranay A, Xu K, LaManna J, and Puchowicz MA
- Subjects
- Animals, Rats, Mice, Interleukin-10 genetics, Interleukin-10 metabolism, Ketone Bodies metabolism, Brain metabolism, Ketosis metabolism, Diet, Ketogenic
- Abstract
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., (© 2022. Springer Nature Switzerland AG.)
- Published
- 2022
- Full Text
- View/download PDF
9. Overcompensation of CoA Trapping by Di(2-ethylhexyl) Phthalate (DEHP) Metabolites in Livers of Wistar Rats.
- Author
-
Hala D, Petersen LH, Huggett DB, Puchowicz MA, Brunengraber H, and Zhang GF
- Subjects
- Animals, Lipid Metabolism physiology, Male, Oxidation-Reduction, PPAR alpha metabolism, Plasticizers metabolism, Rats, Rats, Wistar, Coenzyme A metabolism, Diethylhexyl Phthalate metabolism, Liver metabolism, Phthalic Acids metabolism
- Abstract
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 β-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.- Published
- 2021
- Full Text
- View/download PDF
10. SMAD2 and SMAD3 differentially regulate adiposity and the growth of subcutaneous white adipose tissue.
- Author
-
Kumari R, Irudayam MJ, Al Abdallah Q, Jones TL, Mims TS, Puchowicz MA, Pierre JF, and Brown CW
- Subjects
- 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, Adipogenesis, Adipose Tissue, White cytology, Adiposity, Intra-Abdominal Fat cytology, Smad2 Protein physiology, Smad3 Protein physiology, Subcutaneous Fat cytology
- Abstract
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., (© 2021 Federation of American Societies for Experimental Biology.)
- Published
- 2021
- Full Text
- View/download PDF
11. Novel role of xanthine oxidase-dependent H 2 O 2 production in 12/15-lipoxygenase-mediated de novo lipogenesis, triglyceride biosynthesis and weight gain.
- Author
-
Govatati S, Pichavaram P, Mani AM, Kumar R, Sharma D, Dienel A, Meena S, Puchowicz MA, Park EA, and Rao GN
- Subjects
- Animals, Arachidonate 15-Lipoxygenase genetics, Arachidonate 15-Lipoxygenase metabolism, Diet, High-Fat adverse effects, Hydrogen Peroxide metabolism, Liver metabolism, Mice, Triglycerides metabolism, Weight Gain, Lipogenesis, Xanthine Oxidase metabolism
- Abstract
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 H
2 O2 . 12/15-LOX deficiency leads to cullin2-mediated ubiquitination and degradation of XO, thereby suppressing H2 O2 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., (Copyright © 2021 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
12. Endothelial Thioredoxin-Interacting Protein Depletion Reduces Hemorrhagic Transformation in Hyperglycemic Mice after Embolic Stroke and Thrombolytic Therapy.
- Author
-
Salman M, Ismael S, Li L, Ahmed HA, Puchowicz MA, and Ishrat T
- Abstract
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.
- Published
- 2021
- Full Text
- View/download PDF
13. Chronic Ketosis Modulates HIF1α-Mediated Inflammatory Response in Rat Brain.
- Author
-
Sethuraman A, Rao P, Pranay A, Xu K, LaManna JC, and Puchowicz MA
- Subjects
- Animals, Brain, Ketone Bodies, Mice, Rats, Brain Ischemia, Diet, Ketogenic, Ketosis
- Abstract
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.
- Published
- 2021
- Full Text
- View/download PDF
14. Myeloid Slc2a1 -Deficient Murine Model Revealed Macrophage Activation and Metabolic Phenotype Are Fueled by GLUT1.
- Author
-
Freemerman AJ, Zhao L, Pingili AK, Teng B, Cozzo AJ, Fuller AM, Johnson AR, Milner JJ, Lim MF, Galanko JA, Beck MA, Bear JE, Rotty JD, Bezavada L, Smallwood HS, Puchowicz MA, Liu J, Locasale JW, Lee DP, Bennett BJ, Abel ED, Rathmell JC, and Makowski L
- Subjects
- Animals, Glucose Transporter Type 1 deficiency, Mice, Mice, Inbred C57BL, Mice, Knockout, Phenotype, Disease Models, Animal, Glucose Transporter Type 1 metabolism, Macrophages metabolism
- Abstract
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 Slc2a1
M-/- 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., (Copyright © 2019 by The American Association of Immunologists, Inc.)- Published
- 2019
- Full Text
- View/download PDF
15. Disruption of brain-derived neurotrophic factor production from individual promoters generates distinct body composition phenotypes in mice.
- Author
-
McAllan L, Maynard KR, Kardian AS, Stayton AS, Fox SL, Stephenson EJ, Kinney CE, Alshibli NK, Gomes CK, Pierre JF, Puchowicz MA, Bridges D, Martinowich K, and Han JC
- Subjects
- Animals, Brain-Derived Neurotrophic Factor metabolism, Calorimetry, Indirect, Eating genetics, Energy Metabolism genetics, Mice, Mice, Transgenic, Obesity metabolism, Phenotype, Body Composition genetics, Body Weight genetics, Brain-Derived Neurotrophic Factor genetics, Obesity genetics, Promoter Regions, Genetic
- Abstract
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.- Published
- 2018
- Full Text
- View/download PDF
16. Macrophages with a deletion of the phosphoenolpyruvate carboxykinase 1 ( Pck1 ) gene have a more proinflammatory phenotype.
- Author
-
Ko CW, Counihan D, Wu J, Hatzoglou M, Puchowicz MA, and Croniger CM
- Subjects
- 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, Gene Deletion, Macrophages enzymology, Phenotype, Phosphoenolpyruvate Carboxykinase (GTP) deficiency, Phosphoenolpyruvate Carboxykinase (GTP) genetics
- Abstract
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-
13 C]glucose revealed reduced13 C labeling of citrate and malate and increased13 C 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-1β, 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., (© 2018 by The American Society for Biochemistry and Molecular Biology, Inc.)- Published
- 2018
- Full Text
- View/download PDF
17. Post-resuscitation Arterial Blood Pressure on Survival and Change of Capillary Density Following Cardiac Arrest and Resuscitation in Rats.
- Author
-
Xu K, Puchowicz MA, and LaManna JC
- Subjects
- 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, Brain blood supply, Brain Ischemia etiology, Heart Arrest complications, Hypotension etiology
- Abstract
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/mm
2 ) 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.- Published
- 2018
- Full Text
- View/download PDF
18. Novel ketone body therapy for managing Alzheimer's disease: An Editorial Highlight for 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.
- Author
-
Puchowicz MA and Seyfried TN
- Subjects
- Amino Acids, Animals, Hippocampus, Humans, Mice, Mice, Transgenic, Alzheimer Disease, Citric Acid Cycle
- Abstract
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., (© 2017 International Society for Neurochemistry.)
- Published
- 2017
- Full Text
- View/download PDF
19. Diet-Induced Ketosis Protects Against Focal Cerebral Ischemia in Mouse.
- Author
-
Xu K, Ye L, Sharma K, Jin Y, Harrison MM, Caldwell T, Berthiaume JM, Luo Y, LaManna JC, and Puchowicz MA
- Subjects
- 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, Brain Ischemia prevention & control, Diet, Ketogenic, Infarction, Middle Cerebral Artery diet therapy, Ketosis pathology
- Abstract
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 mm
3 , 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.- Published
- 2017
- Full Text
- View/download PDF
20. Aging Effect on Post-recovery Hypofusion and Mortality Following Cardiac Arrest and Resuscitation in Rats.
- Author
-
Xu K, Puchowicz MA, and LaManna JC
- Subjects
- Animals, Cerebrovascular Circulation, Heart Arrest physiopathology, Male, Rats, Rats, Inbred F344, Reperfusion Injury etiology, Aging physiology, Cardiopulmonary Resuscitation, Heart Arrest mortality
- Abstract
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.
- Published
- 2016
- Full Text
- View/download PDF
21. Protective Effect of Dl-3-n-Butylphthalide on Recovery from Cardiac Arrest and Resuscitation in Rats.
- Author
-
Zhang L, Puchowicz MA, LaManna JC, and Xu K
- Subjects
- 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, Benzofurans pharmacology, CA1 Region, Hippocampal drug effects, Heart Arrest therapy, Neurons drug effects, Neuroprotective Agents pharmacology, Resuscitation
- Abstract
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.
- Published
- 2016
- Full Text
- View/download PDF
22. Decreased carbon shunting from glucose toward oxidative metabolism in diet-induced ketotic rat brain.
- Author
-
Zhang Y, Zhang S, Marin-Valencia I, and Puchowicz MA
- Subjects
- Acetoacetates metabolism, Animals, Energy Metabolism, Ketone Bodies metabolism, Male, Oxidation-Reduction, Rats, Rats, Wistar, gamma-Aminobutyric Acid biosynthesis, Brain Chemistry, Carbon metabolism, Diet, Ketogenic, Glucose metabolism, Propionic Acidemia metabolism
- Abstract
The mechanistic link of ketosis to neuroprotection under certain pathological conditions continues to be explored. We investigated whether chronic ketosis induced by ketogenic diet results in the partitioning of ketone bodies toward oxidative metabolism in brain. We hypothesized that diet-induced ketosis results in increased shunting of ketone bodies toward citric acid cycle and amino acids with decreased carbon shunting from glucose. Rats were fed standard (STD) or ketogenic (KG) diets for 3.5 weeks and then infused with [U-(13) C]glucose or [U-(13) C]acetoacetate tracers. Concentrations and (13) C-labeling pattern of citric acid cycle intermediates and amino acids were analyzed from brain homogenates using stable isotopomer mass spectrometry analysis. The contribution of [U-(13) C]glucose to acetyl-CoA and amino acids decreased by ~ 30% in the KG group versus STD, whereas [U-(13) C]acetoacetate contributions were more than two-fold higher. The concentration of GABA remained constant across groups; however, the (13) C labeling of GABA was markedly increased in the KG group infused with [U-(13) C]acetoacetate compared to STD. This study reveals that there is a significant contribution of ketone bodies to oxidative metabolism and GABA in diet-induced ketosis. We propose that this represents a fundamental mechanism of neuroprotection under pathological conditions., (© 2014 International Society for Neurochemistry.)
- Published
- 2015
- Full Text
- View/download PDF
23. Increasing NAD synthesis in muscle via nicotinamide phosphoribosyltransferase is not sufficient to promote oxidative metabolism.
- Author
-
Frederick DW, Davis JG, Dávila A Jr, Agarwal B, Michan S, Puchowicz MA, Nakamaru-Ogiso E, and Baur JA
- Subjects
- Animals, Binding Sites, Calorimetry, Chromatography, High Pressure Liquid, Mice, Mice, Inbred C57BL, Mice, Transgenic, Mitochondria, Muscle metabolism, NAD metabolism, Nicotinamide Mononucleotide metabolism, Oxidation-Reduction, Poly(ADP-ribose) Polymerases metabolism, Cytokines metabolism, Muscle, Skeletal metabolism, NAD biosynthesis, Nicotinamide Phosphoribosyltransferase metabolism, Oxygen metabolism
- Abstract
The NAD biosynthetic precursors nicotinamide mononucleotide and nicotinamide riboside are reported to confer resistance to metabolic defects induced by high fat feeding in part by promoting oxidative metabolism in skeletal muscle. Similar effects are obtained by germ line deletion of major NAD-consuming enzymes, suggesting that the bioavailability of NAD is limiting for maximal oxidative capacity. However, because of their systemic nature, the degree to which these interventions exert cell- or tissue-autonomous effects is unclear. Here, we report a tissue-specific approach to increase NAD biosynthesis only in muscle by overexpressing nicotinamide phosphoribosyltransferase, the rate-limiting enzyme in the salvage pathway that converts nicotinamide to NAD (mNAMPT mice). These mice display a ∼50% increase in skeletal muscle NAD levels, comparable with the effects of dietary NAD precursors, exercise regimens, or loss of poly(ADP-ribose) polymerases yet surprisingly do not exhibit changes in muscle mitochondrial biogenesis or mitochondrial function and are equally susceptible to the metabolic consequences of high fat feeding. We further report that chronic elevation of muscle NAD in vivo does not perturb the NAD/NADH redox ratio. These studies reveal for the first time the metabolic effects of tissue-specific increases in NAD synthesis and suggest that critical sites of action for supplemental NAD precursors reside outside of the heart and skeletal muscle., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
24. Catabolism of (2E)-4-hydroxy-2-nonenal via ω- and ω-1-oxidation stimulated by ketogenic diet.
- Author
-
Jin Z, Berthiaume JM, Li Q, Henry F, Huang Z, Sadhukhan S, Gao P, Tochtrop GP, Puchowicz MA, and Zhang GF
- Subjects
- Acyl Coenzyme A metabolism, Animals, Brain metabolism, Cytochrome P-450 CYP4A metabolism, Fatty Acids chemistry, Fatty Acids metabolism, Hydroxylation, Kidney metabolism, Lipid Peroxidation, Male, Mass Spectrometry, Metabolism, Myocardium metabolism, Oxidative Stress, Perfusion, Rats, Rats, Sprague-Dawley, Aldehydes metabolism, Diet, Ketogenic, Liver enzymology, Oxygen metabolism
- Abstract
Oxidative stress triggers the peroxidation of ω-6-polyunsaturated fatty acids to reactive lipid fragments, including (2E)-4-hydroxy-2-nonenal (HNE). We previously reported two parallel catabolic pathways of HNE. In this study, we report a novel metabolite that accumulates in rat liver perfused with HNE or 4-hydroxynonanoic acid (HNA), identified as 3-(5-oxotetrahydro-2-furanyl)propanoyl-CoA. In experiments using a combination of isotopic analysis and metabolomics studies, three catabolic pathways of HNE were delineated following HNE conversion to HNA. (i) HNA is ω-hydroxylated to 4,9-dihydroxynonanoic acid, which is subsequently oxidized to 4-hydroxynonanedioic acid. This is followed by the degradation of 4-hydroxynonanedioic acid via β-oxidation originating from C-9 of HNA breaking down to 4-hydroxynonanedioyl-CoA, 4-hydroxyheptanedioyl-CoA, or its lactone, 2-hydroxyglutaryl-CoA, and 2-ketoglutaric acid entering the citric acid cycle. (ii) ω-1-hydroxylation of HNA leads to 4,8-dihydroxynonanoic acid (4,8-DHNA), which is subsequently catabolized via two parallel pathways we previously reported. In catabolic pathway A, 4,8-DHNA is catabolized to 4-phospho-8-hydroxynonanoyl-CoA, 3,8-dihydroxynonanoyl-CoA, 6-hydroxyheptanoyl-CoA, 4-hydroxypentanoyl-CoA, propionyl-CoA, and acetyl-CoA. (iii) The catabolic pathway B of 4,8-DHNA leads to 2,6-dihydroxyheptanoyl-CoA, 5-hydroxyhexanoyl-CoA, 3-hydroxybutyryl-CoA, and acetyl-CoA. Both in vivo and in vitro experiments showed that HNE can be catabolically disposed via ω- and ω-1-oxidation in rat liver and kidney, with little activity in brain and heart. Dietary experiments showed that ω- and ω-1-hydroxylation of HNA in rat liver were dramatically up-regulated by a ketogenic diet, which lowered HNE basal level. HET0016 inhibition and mRNA expression level suggested that the cytochrome P450 4A are main enzymes responsible for the NADPH-dependent ω- and ω-1-hydroxylation of HNA/HNE., (© 2014 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
25. Synergistic simvastatin and metformin combination chemotherapy for osseous metastatic castration-resistant prostate cancer.
- Author
-
Babcook MA, Shukla S, Fu P, Vazquez EJ, Puchowicz MA, Molter JP, Oak CZ, MacLennan GT, Flask CA, Lindner DJ, Parker Y, Daneshgari F, and Gupta S
- Subjects
- Animals, Anticholesteremic Agents administration & dosage, Anticholesteremic Agents pharmacology, Cell Movement drug effects, Drug Synergism, Humans, Hypoglycemic Agents administration & dosage, Hypoglycemic Agents pharmacology, Male, Metformin administration & dosage, Mice, Neoplasm Metastasis, Prostatic Neoplasms, Castration-Resistant pathology, Simvastatin administration & dosage, Antineoplastic Combined Chemotherapy Protocols pharmacology, Bone Neoplasms drug therapy, Bone Neoplasms secondary, Metformin pharmacology, Prostatic Neoplasms, Castration-Resistant drug therapy, Simvastatin pharmacology
- Abstract
Docetaxel chemotherapy remains a standard of care for metastatic castration-resistant prostate cancer (CRPC). Docetaxel modestly increases survival, yet results in frequent occurrence of side effects and resistant disease. An alternate chemotherapy with greater efficacy and minimal side effects is needed. Acquisition of metabolic aberrations promoting increased survival and metastasis in CRPC cells includes constitutive activation of Akt, loss of adenosine monophosphate-activated protein kinase (AMPK) activity due to Ser-485/491 phosphorylation, and overexpression of 3-hydroxy-3-methylglutaryl-Coenzyme A reductase (HMG-CoAR). We report that combination of simvastatin and metformin, within pharmacologic dose range (500 nmol/L to 4 μmol/L simvastatin and 250 μmol/L to 2 mmol/L metformin), significantly and synergistically reduces C4-2B3/B4 CRPC cell viability and metastatic properties, with minimal adverse effects on normal prostate epithelial cells. Combination of simvastatin and metformin decreased Akt Ser-473 and Thr-308 phosphorylation and AMPKα Ser-485/491 phosphorylation; increased Thr-172 phosphorylation and AMPKα activity, as assessed by increased Ser-79 and Ser-872 phosphorylation of acetyl-CoA carboxylase and HMG-CoAR, respectively; decreased HMG-CoAR activity; and reduced total cellular cholesterol and its synthesis in both cell lines. Studies of C4-2B4 orthotopic NCr-nu/nu mice further demonstrated that combination of simvastatin and metformin (3.5-7.0 μg/g body weight simvastatin and 175-350 μg/g body weight metformin) daily by oral gavage over a 9-week period significantly inhibited primary ventral prostate tumor formation, cachexia, bone metastasis, and biochemical failure more effectively than 24 μg/g body weight docetaxel intraperitoneally injected every 3 weeks, 7.0 μg/g/day simvastatin, or 350 μg/g/day metformin treatment alone, with significantly less toxicity and mortality than docetaxel, establishing combination of simvastatin and metformin as a promising chemotherapeutic alternative for metastatic CRPC., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
26. Lipotoxic disruption of NHE1 interaction with PI(4,5)P2 expedites proximal tubule apoptosis.
- Author
-
Khan S, Abu Jawdeh BG, Goel M, Schilling WP, Parker MD, Puchowicz MA, Yadav SP, Harris RC, El-Meanawy A, Hoshi M, Shinlapawittayatorn K, Deschênes I, Ficker E, and Schelling JR
- Subjects
- Acyl Coenzyme A metabolism, Animals, Binding, Competitive, Cation Transport Proteins chemistry, Diabetic Nephropathies etiology, Diabetic Nephropathies pathology, Kidney metabolism, Kidney pathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Obese, Nitric Oxide Synthase Type III deficiency, Nitric Oxide Synthase Type III genetics, Phosphatidylinositol 4,5-Diphosphate chemistry, Protein Binding, Renal Insufficiency, Chronic etiology, Renal Insufficiency, Chronic pathology, Sodium-Hydrogen Exchanger 1, Sodium-Hydrogen Exchangers chemistry, Apoptosis, Cation Transport Proteins metabolism, Kidney Tubules, Proximal physiology, Phosphatidylinositol 4,5-Diphosphate metabolism, Sodium-Hydrogen Exchangers metabolism
- Abstract
Chronic kidney disease progression can be predicted based on the degree of tubular atrophy, which is the result of proximal tubule apoptosis. The Na+/H+ exchanger NHE1 regulates proximal tubule cell survival through interaction with phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2], but pathophysiologic triggers for NHE1 inactivation are unknown. Because glomerular injury permits proximal tubule luminal exposure and reabsorption of fatty acid/albumin complexes, we hypothesized that accumulation of amphipathic, long-chain acyl-CoA (LC-CoA) metabolites stimulates lipoapoptosis by competing with the structurally similar PI(4,5)P2 for NHE1 binding. Kidneys from mouse models of progressive, albuminuric kidney disease exhibited increased fatty acids, LC-CoAs, and caspase-2-dependent proximal tubule lipoapoptosis. LC-CoAs and the cytosolic domain of NHE1 directly interacted, with an affinity comparable to that of the PI(4,5)P2-NHE1 interaction, and competing LC-CoAs disrupted binding of the NHE1 cytosolic tail to PI(4,5)P2. Inhibition of LC-CoA catabolism reduced NHE1 activity and enhanced apoptosis, whereas inhibition of proximal tubule LC-CoA generation preserved NHE1 activity and protected against apoptosis. Our data indicate that albuminuria/lipiduria enhances lipotoxin delivery to the proximal tubule and accumulation of LC-CoAs contributes to tubular atrophy by severing the NHE1-PI(4,5)P2 interaction, thereby lowering the apoptotic threshold. Furthermore, these data suggest that NHE1 functions as a metabolic sensor for lipotoxicity.
- Published
- 2014
- Full Text
- View/download PDF
27. Sterol regulatory element-binding protein-1 (SREBP-1) is required to regulate glycogen synthesis and gluconeogenic gene expression in mouse liver.
- Author
-
Ruiz R, Jideonwo V, Ahn M, Surendran S, Tagliabracci VS, Hou Y, Gamble A, Kerner J, Irimia-Dominguez JM, Puchowicz MA, DePaoli-Roach A, Hoppel C, Roach P, and Morral N
- Subjects
- Animals, Diabetes Mellitus, Type 1 genetics, Diabetes Mellitus, Type 1 metabolism, Diabetes Mellitus, Type 1 pathology, Diabetes Mellitus, Type 2 genetics, Diabetes Mellitus, Type 2 metabolism, Diabetes Mellitus, Type 2 pathology, Gene Knockdown Techniques, Glycogen genetics, Male, Mice, Obesity genetics, Obesity metabolism, Obesity pathology, Sterol Regulatory Element Binding Protein 1 genetics, Gene Expression Regulation physiology, Gluconeogenesis physiology, Glycogen biosynthesis, Liver metabolism, Sterol Regulatory Element Binding Protein 1 metabolism
- Abstract
Sterol regulatory element-binding protein-1 (SREBP-1) is a key transcription factor that regulates genes in the de novo lipogenesis and glycolysis pathways. The levels of SREBP-1 are significantly elevated in obese patients and in animal models of obesity and type 2 diabetes, and a vast number of studies have implicated this transcription factor as a contributor to hepatic lipid accumulation and insulin resistance. However, its role in regulating carbohydrate metabolism is poorly understood. Here we have addressed whether SREBP-1 is needed for regulating glucose homeostasis. Using RNAi and a new generation of adenoviral vector, we have silenced hepatic SREBP-1 in normal and obese mice. In normal animals, SREBP-1 deficiency increased Pck1 and reduced glycogen deposition during fed conditions, providing evidence that SREBP-1 is necessary to regulate carbohydrate metabolism during the fed state. Knocking SREBP-1 down in db/db mice resulted in a significant reduction in triglyceride accumulation, as anticipated. However, mice remained hyperglycemic, which was associated with up-regulation of gluconeogenesis gene expression as well as decreased glycolysis and glycogen synthesis gene expression. Furthermore, glycogen synthase activity and glycogen accumulation were significantly reduced. In conclusion, silencing both isoforms of SREBP-1 leads to significant changes in carbohydrate metabolism and does not improve insulin resistance despite reducing steatosis in an animal model of obesity and type 2 diabetes.
- Published
- 2014
- Full Text
- View/download PDF
28. Ketosis proportionately spares glucose utilization in brain.
- Author
-
Zhang Y, Kuang Y, Xu K, Harris D, Lee Z, LaManna J, and Puchowicz MA
- Subjects
- Algorithms, Animals, Blood Glucose metabolism, Diet, Diet, Ketogenic, Fluorodeoxyglucose F18, Image Processing, Computer-Assisted, Ketone Bodies metabolism, Lactic Acid metabolism, Male, Positron-Emission Tomography, Radiopharmaceuticals, Rats, Rats, Wistar, Brain metabolism, Glucose metabolism, Ketosis metabolism
- Abstract
The brain is dependent on glucose as a primary energy substrate, but is capable of utilizing ketones such as β-hydroxybutyrate and acetoacetate, as occurs with fasting, starvation, or chronic feeding of a ketogenic diet. The relationship between changes in cerebral metabolic rates of glucose (CMRglc) and degree or duration of ketosis remains uncertain. To investigate if CMRglc decreases with chronic ketosis, 2-[(18)F]fluoro-2-deoxy-D-glucose in combination with positron emission tomography, was applied in anesthetized young adult rats fed 3 weeks of either standard or ketogenic diets. Cerebral metabolic rates of glucose (μmol/min per 100 g) was determined in the cerebral cortex and cerebellum using Gjedde-Patlak analysis. The average CMRglc significantly decreased in the cerebral cortex (23.0±4.9 versus 32.9±4.7) and cerebellum (29.3±8.6 versus 41.2±6.4) with increased plasma ketone bodies in the ketotic rats compared with standard diet group. The reduction of CMRglc in both brain regions correlates linearly by ∼9% for each 1 mmol/L increase of total plasma ketone bodies (0.3 to 6.3 mmol/L). Together with our meta-analysis, these data revealed that the degree and duration of ketosis has a major role in determining the corresponding change in CMRglc with ketosis.
- Published
- 2013
- Full Text
- View/download PDF
29. Targeting pyruvate carboxylase reduces gluconeogenesis and adiposity and improves insulin resistance.
- Author
-
Kumashiro N, Beddow SA, Vatner DF, Majumdar SK, Cantley JL, Guebre-Egziabher F, Fat I, Guigni B, Jurczak MJ, Birkenfeld AL, Kahn M, Perler BK, Puchowicz MA, Manchem VP, Bhanot S, Still CD, Gerhard GS, Petersen KF, Cline GW, Shulman GI, and Samuel VT
- Subjects
- Adipose Tissue enzymology, Adult, Animals, Fatty Liver enzymology, Female, Glycerol metabolism, Humans, Male, Middle Aged, Rats, Rats, Sprague-Dawley, Rats, Zucker, Adiposity physiology, Gluconeogenesis physiology, Insulin Resistance physiology, Liver enzymology, Pyruvate Carboxylase metabolism
- Abstract
We measured the mRNA and protein expression of the key gluconeogenic enzymes in human liver biopsy specimens and found that only hepatic pyruvate carboxylase protein levels related strongly with glycemia. We assessed the role of pyruvate carboxylase in regulating glucose and lipid metabolism in rats through a loss-of-function approach using a specific antisense oligonucleotide (ASO) to decrease expression predominantly in liver and adipose tissue. Pyruvate carboxylase ASO reduced plasma glucose concentrations and the rate of endogenous glucose production in vivo. Interestingly, pyruvate carboxylase ASO also reduced adiposity, plasma lipid concentrations, and hepatic steatosis in high fat-fed rats and improved hepatic insulin sensitivity. Pyruvate carboxylase ASO had similar effects in Zucker Diabetic Fatty rats. Pyruvate carboxylase ASO did not alter de novo fatty acid synthesis, lipolysis, or hepatocyte fatty acid oxidation. In contrast, the lipid phenotype was attributed to a decrease in hepatic and adipose glycerol synthesis, which is important for fatty acid esterification when dietary fat is in excess. Tissue-specific inhibition of pyruvate carboxylase is a potential therapeutic approach for nonalcoholic fatty liver disease, hepatic insulin resistance, and type 2 diabetes.
- Published
- 2013
- Full Text
- View/download PDF
30. Metabolomics and mass isotopomer analysis as a strategy for pathway discovery: pyrrolyl and cyclopentenyl derivatives of the pro-drug of abuse, levulinate.
- Author
-
Harris SR, Zhang GF, Sadhukhan S, Wang H, Shi C, Puchowicz MA, Anderson VE, Salomon RG, Tochtrop GP, and Brunengraber H
- Subjects
- Animals, Brain metabolism, Coenzyme A chemistry, Cyclization, Enzyme Inhibitors chemistry, Levulinic Acids chemistry, Liver metabolism, Male, Metabolomics, Prodrugs chemistry, Rats, Rats, Sprague-Dawley, Coenzyme A metabolism, Enzyme Inhibitors metabolism, Levulinic Acids metabolism, Prodrugs metabolism
- Abstract
We recently reported that levulinate (4-ketopentanoate) is converted in the liver to 4-hydroxypentanoate, a drug of abuse, and that the formation of 4-hydroxypentanoate is stimulated by ethanol oxidation. We also identified 3 parallel β-oxidation pathways by which levulinate and 4-hydroxypentanoate are catabolized to propionyl-CoA and acetyl-CoA. We now report that levulinate forms three seven-carbon cyclical CoA esters by processes starting with the elongation of levulinyl-CoA by acetyl-CoA to 3,6-diketoheptanoyl-CoA. The latter γ-diketo CoA ester undergoes two parallel cyclization processes. One process yields a mixture of tautomers, i.e., cyclopentenyl- and cyclopentadienyl-acyl-CoAs. The second cyclization process yields a methyl-pyrrolyl-acetyl-CoA containing a nitrogen atom derived from the ε-nitrogen of lysine but without carbons from lysine. The cyclic CoA esters were identified in rat livers perfused with levulinate and in livers and brains from rats gavaged with calcium levulinate ± ethanol. Lastly, 3,6-diketoheptanoyl-CoA, like 2,5-diketohexane, pyrrolates free lysine and, presumably, lysine residues from proteins. This may represent a new pathway for protein pyrrolation. The cyclic CoA esters and related pyrrolation processes may play a role in the toxic effects of 4-hydroxypentanoate.
- Published
- 2013
- Full Text
- View/download PDF
31. Contribution of brain glucose and ketone bodies to oxidative metabolism.
- Author
-
Zhang Y, Kuang Y, LaManna JC, and Puchowicz MA
- Subjects
- Animals, Carbon Radioisotopes, Chromatography, Liquid, Diet, Ketogenic, Gas Chromatography-Mass Spectrometry, Male, Oxidation-Reduction, Rats, Rats, Wistar, Acetoacetates metabolism, Acetyl Coenzyme A metabolism, Brain metabolism, Glucose chemistry, Glucose metabolism, Ketone Bodies chemistry, Ketone Bodies metabolism
- Abstract
Unlabelled: Ketone bodies are an alternative energy substrate to glucose in brain. Under conditions of oxidative stress, we hypothesize that ketosis stabilizes glucose metabolism by partitioning glucose away from oxidative metabolism towards ketone body oxidation. In this study we assessed oxidative metabolism in ketotic rat brain using stable isotope mass spectrometry analysis. The contribution of glucose and ketone bodies to oxidative metabolism was studied in cortical brain homogenates isolated from anesthetized ketotic rats. To induce chronic ketosis, rats were fed either a ketogenic (high-fat, carbohydrate restricted) or standard rodent chow for 3 weeks and then infused intravenously with tracers of [U-(13)C] glucose or [U-(13)C] acetoacetate for 60 min. The measured percent contribution of glucose or ketone bodies to oxidative metabolism was analyzed by measuring the (13)C-label incorporation into acetyl-CoA. Using mass spectrometry (gas-chromatography; GC-MS, and liquid-chromatography; LCMS) and isotopomer analysis, the fractional amount of substrate oxidation was measured as the M + 2 enrichment (%) of acetyl-CoA relative to the achieved enrichment of the infused precursors, [U-(13)C]glucose or [U-(13)C] acetoacetate., Results: the percent contribution of glucose oxidation in cortical brain in rats fed the ketogenic diet was 71.2 ± 16.8 (mean% ± SD) compared to the standard chow, 89.0 ± 14.6. Acetoacetate oxidation was significantly higher with ketosis compared to standard chow, 41.7 ± 9.4 vs. 21.9 ± 10.6. These data confer the high oxidative capacity for glucose irrespective of ketotic or non-ketotic states. With ketosis induced by 3 weeks of diet, cortical brain utilizes twice as much acetoacetate compared to non-ketosis.
- Published
- 2013
- Full Text
- View/download PDF
32. Dietary regulation of catabolic disposal of 4-hydroxynonenal analogs in rat liver.
- Author
-
Li Q, Tomcik K, Zhang S, Puchowicz MA, and Zhang GF
- Subjects
- Aldehydes chemistry, Alkenes chemistry, Alkenes metabolism, Animals, Biomarkers metabolism, Diet, High-Fat adverse effects, Diet, Ketogenic adverse effects, Glutathione chemistry, Glutathione metabolism, Male, Mass Spectrometry, Oxidation-Reduction, Oxidative Stress, Rats, Rats, Wistar, Aldehydes metabolism, Dietary Fats, Unsaturated metabolism, Lipid Peroxidation, Liver metabolism
- Abstract
Our previous work in perfused rat livers has demonstrated that 4-hydroxynonenal (HNE) is catabolized predominantly via β oxidation. Therefore, we hypothesized that perturbations in β oxidation, such as diet-altered fatty acid oxidation activity, could lead to changes in HNE levels. To test our hypothesis, we (i) developed a simple and sensitive GC/MS method combined with mass isotopomer analysis to measure HNE and HNE analogs, 4-oxononenal (ONE) and 1,4-dihydroxynonene (DHN), and (ii) investigated the effects of four diets (standard, low-fat, ketogenic, and high-fat mix) on HNE, ONE, and DHN concentrations in rat livers. Our results showed that livers from rats fed the ketogenic diet or high-fat mix diet had high ω-6 polyunsaturated fatty acid concentrations and markers of oxidative stress. However, high concentrations of HNE (1.6 ± 0.5 nmol/g) and ONE (0.9 ± 0.2 nmol/g) were found only in livers from rats fed the high-fat mix diet. Livers from rats fed the ketogenic diet had low HNE (0.8 ± 0.1 nmol/g) and ONE (0.4 ± 0.07 nmol/g), similar to rats fed the standard diet. A possible explanation is that the predominant pathway of HNE catabolism (i.e., β oxidation) is activated in the liver by the ketogenic diet. This is consistent with a 10-fold decrease in malonyl-CoA in livers from rats fed a ketogenic diet compared to a standard diet. The accelerated catabolism of HNE lowers HNE and HNE analog concentrations in livers from rats fed the ketogenic diet. On the other hand, rats fed the high-fat mix diet had high rates of lipid synthesis and low rates of fatty acid oxidation, resulting in the slowing down of the catabolic disposal of HNE and HNE analogs. Thus, decreased HNE catabolism from a high-fat mix diet induces high concentrations of HNE and HNE analogs. The results of this work suggest a potential causal relationship to metabolic syndrome induced by Western diets (i.e., high-fat mix), as well as the effects of a ketogenic diet on the catabolism of lipid peroxidation products in liver., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
33. Neuroprotective properties of ketone bodies.
- Author
-
Xu K, Lamanna JC, and Puchowicz MA
- Subjects
- Animals, Brain metabolism, Brain Ischemia chemically induced, Glucose metabolism, Heart Arrest chemically induced, Heart Arrest drug therapy, Male, Rats, Rats, Wistar, Stroke chemically induced, Brain blood supply, Brain Ischemia drug therapy, Energy Metabolism, Ketone Bodies metabolism, Ketosis, Stroke drug therapy
- Published
- 2012
- Full Text
- View/download PDF
34. Regional brain blood flow in mouse: quantitative measurement using a single-pass radio-tracer method and a mathematical algorithm.
- Author
-
Xu K, Radhakrishnan K, Serhal A, Allen F, Lamanna JC, and Puchowicz MA
- Subjects
- Animals, Female, Male, Mice, Mice, Inbred C57BL, Rats, Algorithms, Brain blood supply, Carbon Radioisotopes, Models, Theoretical, Regional Blood Flow
- Abstract
We have developed a reliable experimental method for measuring local regional cerebral blood flows in anesthetized mice. This method is an extension of the well-established single-pass dual-label indicator method for simultaneously measuring blood flow and glucose influx in rat brains. C57BL6J mice (n = 10) were anesthetized and regional blood flows (ml/min/g) were measured using the radio-tracer method. To test the sensitivity of this method we used a mathematical algorithm to predict the blood flows and compared the two sets of results.Measured regional blood flows between 0.7 and 1.7 ml/min/g were similar to those we have previously reported in the rat. The predicted blood flows using an assumed linearly increasing arterial tracer concentration-versus-time profile (that is, a ramp) were similar to the values measured in the physiological experiments (R(2) 0.99; slope 0.91). Thus,measurements of local regional cerebral blood flow in anesthetized mice using a single-pass radio-tracer method appear to be reliable.
- Published
- 2011
- Full Text
- View/download PDF
35. Analysis of the citric acid cycle intermediates using gas chromatography-mass spectrometry.
- Author
-
Kombu RS, Brunengraber H, and Puchowicz MA
- Subjects
- Chloroform chemistry, Methanol chemistry, Perfusion, Plasma chemistry, Tissue Extracts chemistry, Water chemistry, Citric Acid Cycle, Gas Chromatography-Mass Spectrometry, Metabolomics methods
- Abstract
Researchers view analysis of the citric acid cycle (CAC) intermediates as a metabolomic approach to identifying unexpected correlations between apparently related and unrelated pathways of metabolism. Relationships of the CAC intermediates, as measured by their concentrations and relative ratios, offer useful information to understanding interrelationships between the CAC and metabolic pathways under various physiological and pathological conditions. This chapter presents a relatively simple method that is sensitive for simultaneously measuring concentrations of CAC intermediates (relative and absolute) and other related intermediates of energy metabolism using gas chromatography-mass spectrometry.
- Published
- 2011
- Full Text
- View/download PDF
36. Decreased brainstem function following cardiac arrest and resuscitation in aged rat.
- Author
-
Xu K, Puchowicz MA, Sun X, and LaManna JC
- Subjects
- Animals, Antioxidants pharmacology, Brain Stem pathology, Brain Stem Infarctions etiology, Brain Stem Infarctions pathology, Brain Stem Infarctions physiopathology, Cell Respiration physiology, Cyclic N-Oxides pharmacology, Disease Models, Animal, Hippocampus pathology, Hippocampus physiopathology, Male, Mitochondria metabolism, Mitochondria pathology, Nerve Degeneration etiology, Nerve Degeneration pathology, Nerve Degeneration physiopathology, Oxidative Stress drug effects, Oxidative Stress physiology, Rats, Rats, Inbred F344, Recovery of Function physiology, Reperfusion Injury drug therapy, Reperfusion Injury pathology, Respiratory Insufficiency etiology, Respiratory Insufficiency pathology, Respiratory Insufficiency physiopathology, Treatment Outcome, Aging physiology, Brain Stem physiopathology, Heart Arrest complications, Reperfusion Injury physiopathology, Resuscitation adverse effects
- Abstract
There is a high incidence of cardiac arrest and poorer post-resuscitation outcome in the elderly population. Cardiac arrest and resuscitation results in ischemia/reperfusion injury associated with oxidative stress, leading to post-resuscitation mortality and delayed selective neuronal cell loss. In this study we investigated recovery following cardiac arrest and resuscitation in the aged rat brain. Male Fischer 344 rats (6, 12 and 24 months old) underwent 7 minute cardiac arrest before resuscitation. Overall survival and hippocampal neuronal counts were determined at 4 days of recovery. Brainstem function was assessed by hypoxic ventilatory response (HVR). Mitochondria of brainstem, cortex and hippocampus were isolated and assessed for respiratory function. Effect of an antioxidant, alpha-phenyl-tert-butyl-nitrone (PBN) was used as a treatment strategy against oxidative stress in the 6 and 24-month old rats. The time course of mitochondrial function was established using 3-month old Wistar rats with 12-minute cardiac arrest. In the 24-month old rats, overall survival rate, hippocampal CA1 neuronal counts, HVR, and brain mitochondrial respiratory control ratio were significantly reduced following cardiac arrest and resuscitation compared to the younger rats, and PBN treatment improved outcome. The data suggest that (i) there was increased susceptibility to ischemia/reperfusion in aged rat brain; (ii) HVR was decreased in the aged rats; (iii) brain mitochondrial respiratory function related to coupled oxidation was decreased following cardiac arrest and resuscitation in rats, more so in the aged; and (iv) treatment with an antioxidant, such as PBN, reduced the oxidative damage following cardiac arrest and resuscitation., (Copyright 2010 Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
37. Diet-induced ketosis improves cognitive performance in aged rats.
- Author
-
Xu K, Sun X, Eroku BO, Tsipis CP, Puchowicz MA, and LaManna JC
- Subjects
- Animals, Behavior, Animal, Blotting, Western, Capillaries cytology, Capillaries metabolism, Cell Count, Cerebral Cortex metabolism, Glucose Transporter Type 1 metabolism, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Male, Rats, Rats, Inbred F344, Aging pathology, Cognition, Diet, Ketogenic, Ketosis pathology
- Abstract
Aging is associated with increased susceptibility to hypoxic/ischemic insult and declines in behavioral function which may be due to attenuated adaptive/defense responses. We investigated if diet-induced ketosis would improve behavioral performance in the aged rats. Fischer 344 rats (3- and 22-month-old) were fed standard (STD) or ketogenic (KG) diet for 3 weeks and then exposed to hypobaric hypoxia. Cognitive function was measured using the T-maze and object recognition tests. Motor function was measured using the inclined-screen test. Results showed that KG diet significantly increased blood ketone levels in both young and old rats. In the aged rats, the KG diet improved cognitive performance under normoxic and hypoxic conditions; while motor performance remained unchanged. Capillary density and HIF-1alpha levels were elevated in the aged ketotic group independent of hypoxic challenge. These data suggest that diet-induced ketosis may be beneficial in the treatment of neurodegenerative conditions.
- Published
- 2010
- Full Text
- View/download PDF
38. Neuroprotection in diet-induced ketotic rat brain after focal ischemia.
- Author
-
Puchowicz MA, Zechel JL, Valerio J, Emancipator DS, Xu K, Pundik S, LaManna JC, and Lust WD
- Subjects
- Animals, Brain enzymology, Brain Edema enzymology, Brain Edema metabolism, Brain Infarction enzymology, Brain Infarction metabolism, Brain Ischemia enzymology, Brain Ischemia metabolism, Disease Models, Animal, Hypoxia-Inducible Factor 1, alpha Subunit biosynthesis, Ketosis metabolism, Male, Neuroprotective Agents metabolism, Procollagen-Proline Dioxygenase biosynthesis, Proto-Oncogene Proteins c-bcl-2 biosynthesis, RNA, Messenger biosynthesis, Rats, Rats, Wistar, Reverse Transcriptase Polymerase Chain Reaction, Succinic Acid metabolism, Brain metabolism, Brain Edema prevention & control, Brain Infarction prevention & control, Brain Ischemia diet therapy, Diet, Ketogenic, Ketone Bodies biosynthesis
- Abstract
Neuroprotective properties of ketosis may be related to the upregulation of hypoxia inducible factor (HIF)-1alpha, a primary constituent associated with hypoxic angiogenesis and a regulator of neuroprotective responses. The rationale that the utilization of ketones by the brain results in elevation of intracellular succinate, a known inhibitor of prolyl hydroxylase (the enzyme responsible for the degradation of HIF-1alpha) was deemed as a potential mechanism of ketosis on the upregulation of HIF-1alpha. The neuroprotective effect of diet-induced ketosis (3 weeks of feeding a ketogenic diet), as pretreatment, on infarct volume, after reversible middle cerebral artery occlusion (MCAO), and the upregulation of HIF-1alpha were investigated. The effect of beta-hydroxybutyrate (BHB), as a pretreatment, via intraventricular infusion (4 days of infusion before stroke) was also investigated following MCAO. Levels of HIF-1alpha and Bcl-2 (anti-apoptotic protein) proteins and succinate content were measured. A 55% or 70% reduction in infarct volume was observed with BHB infusion or diet-induced ketosis, respectively. The levels of HIF-1alpha and Bcl-2 proteins increased threefold with diet-induced ketosis; BHB infusions also resulted in increases in these proteins. As hypothesized, succinate content increased by 55% with diet-induced ketosis and fourfold with BHB infusion. In conclusion, the biochemical link between ketosis and the stabilization of HIF-1alpha is through the elevation of succinate, and both HIF-1alpha stabilization and Bcl-2 upregulation play a role in ketone-induced neuroprotection in the brain.
- Published
- 2008
- Full Text
- View/download PDF
39. Mitochondrial dysfunction in aging rat brain following transient global ischemia.
- Author
-
Xu K, Puchowicz MA, Sun X, and LaManna JC
- Subjects
- Aldehydes metabolism, Animals, Brain Ischemia etiology, Brain Ischemia pathology, Cross-Linking Reagents metabolism, Cyclic N-Oxides metabolism, Cyclic N-Oxides pharmacology, Hypoxia, Male, Mitochondria drug effects, Neuroprotective Agents metabolism, Neuroprotective Agents pharmacology, Oxidation-Reduction, Oxygen Consumption, Rats, Rats, Inbred F344, Time Factors, Aging physiology, Brain Ischemia metabolism, Mitochondria metabolism, Oxidative Stress
- Abstract
Aged rat brain is more sensitive to reperfusion injury induced by cardiac arrest and resuscitation. The mitochondrial respiratory chain, the major source of free radicals during reperfusion, is likely to be the target of lipid peroxidation. Previous work has shown a higher mortality and lower hippocampal neuronal survival in older rats. 4-hydroxy-2-nonenal (HNE), a major product of lipid peroxidation, was found to be elevated in cortex and brainstem after resuscitation. In this study we investigated the acute changes of mitochondrial function in aging rat brain following cardiac arrest and resuscitation; the effect of an antioxidant, alpha-phenyl-tert-butyl-nitrone (PBN) was also tested. Fischer 344 rats, 6 and 24-month old, were subjected to cardiac arrest (7-10 minutes) and allowed to recover 1 hour after resuscitation. Mitochondria of cortex and brainstem were isolated and assayed for respiratory function. Compared to their respective non-arrested control group, 1h untreated groups (both 6 month and 24 month) had similar state 3 (ADP-stimulated) but higher state 4 (resting state) respiratory rates. The respiratory control ratio (state 3/state 4) of cortex in the 1h untreated group was 26% lower than the non-arrested control group; similar results were found in brainstem. The decreased mitochondrial respiratory function was improved by PBN treatment. HNE-modified mitochondrial proteins were elevated 1h after resuscitation, with an evident change in the aged. Treatment with PBN reduced the elevated HNE production in mitochondria of cortex. The data suggest (i) there is increased sensitivity to lipid peroxidation with aging, (ii) mitochondrial respiratory function related to coupled oxidation decreases following cardiac arrest and resuscitation, and (iii) treatment with antioxidant, such as PBN, reduces the oxidative damage following cardiac arrest and resuscitation.
- Published
- 2008
- Full Text
- View/download PDF
40. Mutations in the MPV17 gene are responsible for rapidly progressive liver failure in infancy.
- Author
-
Wong LJ, Brunetti-Pierri N, Zhang Q, Yazigi N, Bove KE, Dahms BB, Puchowicz MA, Gonzalez-Gomez I, Schmitt ES, Truong CK, Hoppel CL, Chou PC, Wang J, Baldwin EE, Adams D, Leslie N, Boles RG, Kerr DS, and Craigen WJ
- Subjects
- DNA, Mitochondrial metabolism, Disease Progression, Electron Transport Chain Complex Proteins metabolism, Female, Genetic Testing, Hispanic or Latino ethnology, Hispanic or Latino genetics, Humans, Infant, Infant, Newborn, Liver metabolism, Liver pathology, Liver Failure diagnosis, Liver Failure ethnology, Male, Pedigree, Texas, White People ethnology, White People genetics, Liver Failure genetics, Membrane Proteins genetics, Mutation genetics
- Abstract
Unlabelled: MPV17 is a mitochondrial inner membrane protein of unknown function recently recognized as responsible for a mitochondrial DNA depletion syndrome. The aim of this study is to delineate the specific clinical, pathological, biochemical, and molecular features associated with mitochondrial DNA depletion due to MPV17 gene mutations. We report 4 cases from 3 ethnically diverse families with MPV17 mutations. Importantly, 2 of these cases presented with isolated liver failure during infancy without notable neurologic dysfunction., Conclusion: We therefore propose that mutations in the MPV17 gene be considered in the course of evaluating the molecular etiology for isolated, rapidly progressive infantile hepatic failure.
- Published
- 2007
- Full Text
- View/download PDF
41. Statistical analysis of metabolic pathways of brain metabolism at steady state.
- Author
-
Occhipinti R, Puchowicz MA, LaManna JC, Somersalo E, and Calvetti D
- Subjects
- Animals, Computer Simulation, Data Interpretation, Statistical, Humans, Markov Chains, Models, Statistical, Multienzyme Complexes chemistry, Brain metabolism, Energy Metabolism physiology, Models, Neurological, Nerve Tissue Proteins metabolism, Signal Transduction physiology
- Abstract
The estimation of metabolic fluxes for brain metabolism is important, among other things, to test the validity of different hypotheses which have been proposed in the literature. The metabolic model that we propose considers, in addition to the blood compartment, the cytosol, and mitochondria of both astrocyte and neuron, including detailed metabolic pathways. In this work we use a recently developed methodology to perform a statistical Flux Balance Analysis (FBA) for this model. The methodology recasts the problem in the form of Bayesian statistical inference and therefore can take advantage of qualitative information about brain metabolism for the simultaneous estimation of all reaction fluxes and transport rates at steady state. By a Markov Chain Monte Carlo (MCMC) sampling method, we are able to provide for each reaction flux and transport rate a distribution of possible values. The analysis of the histograms of the reaction fluxes and transport rates provides a very useful tool for assessing the validity of different hypotheses about brain energetics proposed in the literature, and facilitates the design of the pathways network that is in accordance with what is understood of the functioning of the brain. In this work, we focus on the analysis of biochemical pathways within each cell type (astrocyte and neuron) at different levels of neural activity, and we demonstrate how statistical tools can help implement various bounds suggested by experimental data.
- Published
- 2007
- Full Text
- View/download PDF
42. Diet-induced ketosis increases capillary density without altered blood flow in rat brain.
- Author
-
Puchowicz MA, Xu K, Sun X, Ivy A, Emancipator D, and LaManna JC
- Subjects
- 3-Hydroxybutyric Acid blood, 3-Hydroxybutyric Acid metabolism, Animals, Blood Vessels metabolism, Blood-Brain Barrier, Brain metabolism, Capillaries metabolism, Capillaries pathology, Glucose Transporter Type 1 metabolism, Ketosis etiology, Male, Microcirculation, Monocarboxylic Acid Transporters metabolism, Osmolar Concentration, Rats, Rats, Wistar, Symporters metabolism, Tissue Distribution, Brain blood supply, Cerebrovascular Circulation, Diet, Ketosis pathology, Ketosis physiopathology
- Abstract
It is recognized that ketone bodies, such as R-beta-hydroxybutyrate (beta-HB) and acetoacetate, are energy sources for the brain. As with glucose metabolism, monocarboxylate uptake by the brain is dependent on the function and regulation of its own transporter system. We concurrently investigated ketone body influx, blood flow, and regulation of monocarboxylate transporter (MCT-1) and glucose transporter (GLUT-1) in diet-induced ketotic (KG) rat brain. Regional blood-to-brain beta-HB influx (micromol.g(-1).min(-1)) increased 40-fold with ketosis (4.8 +/- 1.8 plasmabeta-HB; mM) in all regions compared with the nonketotic groups (standard and no-fat diets); there were no changes in regional blood flow. Immunohistochemical staining revealed that GLUT-1 density (number/mm2) in the cortex was significantly elevated (40%) in the ketotic group compared with the standard and no-fat diet groups. MCT-1 was also markedly (3-fold) upregulated in the ketotic group compared with the standard diet group. In the standard diet group, 40% of the brain capillaries stained positive for MCT-1; this amount doubled with the ketotic diet. Western blot analysis of isolated microvessels from ketotic rat brain showed an eightfold increase in GLUT-1 and a threefold increase in MCT-1 compared with the standard diet group. These data suggest that diet-induced ketosis results in increased vascular density at the blood-brain barrier without changes in blood flow. The increase in extraction fraction and capillary density with increased plasma ketone bodies indicates a significant flux of substrates available for brain energy metabolism.
- Published
- 2007
- Full Text
- View/download PDF
43. Increased sensitivity to transient global ischemia in aging rat brain.
- Author
-
Xu K, Sun X, Puchowicz MA, and LaManna JC
- Subjects
- Aldehydes metabolism, Animals, Antioxidants metabolism, Brain Ischemia pathology, Cross-Linking Reagents metabolism, Cyclic N-Oxides metabolism, Free Radical Scavengers metabolism, Hippocampus cytology, Hypoxia, Male, Neurons cytology, Neurons metabolism, Neuroprotective Agents metabolism, Rats, Rats, Inbred F344, Survival Rate, Aging physiology, Brain Ischemia metabolism, Oxidative Stress
- Abstract
Transient global brain ischemia induced by cardiac arrest and resuscitation (CAR) results in reperfusion injury associated with oxidative stress. Oxidative stress is known to produce delayed selective neuronal cell loss and impairment of brainstem function, leading to post-resuscitation mortality. Levels of 4-hydroxy-2-nonenal (HNE) modified protein adducts, a marker of oxidative stress, was found to be elevated after CAR in rat brain. In this study we investigated the effects of an antioxidant, alpha-phenyl-tert-butyl-nitrone (PBN) on the recovery following CAR in the aged rat brain. Male Fischer 344 rats (6, 12 and 24-month old) underwent 7-minute cardiac arrest before resuscitation. Brainstem function was assessed by hypoxic ventilatory response (HVR) and HNE-adducts were measured by western blot analysis. Our data showed that in the 24-month old rats, overall survival rate, hippocampal CAl neuronal counts and HVR were significantly reduced compared to the younger rats. With PBN treatment, the recovery was improved in the aged rat brain, which was consistent with reduced HNE adducts in brain following CAR. Our data suggest that aged rats are more vulnerable to oxidative stress insult and treatment with PBN improves the outcome following reperfusion injury. The mechanism of action is most likely through the scavenging of reactive oxygen species resulting in reduced lipid peroxidation.
- Published
- 2007
- Full Text
- View/download PDF
44. Adenosine treatment delays postischemic hippocampal CA1 loss after cardiac arrest and resuscitation in rats.
- Author
-
Xu K, Puchowicz MA, Lust WD, and LaManna JC
- Subjects
- Analysis of Variance, Animals, Body Temperature drug effects, Body Temperature physiology, Brain Edema etiology, Brain Edema pathology, Brain Edema prevention & control, Brain Ischemia drug therapy, Brain Ischemia etiology, Cell Count methods, Cell Death drug effects, Cell Death physiology, Disease Models, Animal, Heart Arrest chemically induced, Heart Arrest complications, Heart Arrest pathology, Male, Neurons physiology, Physical Examination methods, Rats, Rats, Wistar, Recovery of Function drug effects, Recovery of Function physiology, Regional Blood Flow drug effects, Regional Blood Flow physiology, Reperfusion methods, Time Factors, Water metabolism, Adenosine therapeutic use, Anti-Arrhythmia Agents therapeutic use, Heart Arrest therapy, Hippocampus pathology, Neurons drug effects, Resuscitation methods
- Abstract
Resuscitation from cardiac arrest results in reperfusion injury that leads to increased postresuscitation mortality and delayed neuronal death. One of the many consequences of resuscitation from cardiac arrest is a derangement of energy metabolism and the loss of adenylates, impairing the tissue's ability to regain proper energy balance. In this study, we investigated the effects of adenosine (ADO) on the recovery of the brain from 12 min of ischemia using a rat model of cardiac arrest and resuscitation. Compared to the untreated group, treatment with adenosine (7.2 mg/kg) initiated immediately after resuscitation increased the proportion of rats surviving to 4 days and significantly delayed hippocampal CA1 neuronal loss. Brain blood flow was increased significantly in the adenosine-treated rats 1 h after cardiac arrest and resuscitation. Adenosine-treated rats exhibited less edema in cortex, brainstem and hippocampus during the first 48 h of recovery. Adenosine treatment significantly lowered brain temperature during recovery, and a part of the neuroprotective effects of adenosine treatment could be ascribed to adenosine-induced hypothermia. With this dose, adenosine may have a delayed transient effect on the restoration of the adenylate pool (AXP = ATP + ADP + AMP) 24 h after cardiac arrest and resuscitation. Our findings suggested that improved postischemic brain blood flow and ADO-induced hypothermia, rather than adenylate supplementation, may be the two major contributors to the neuroprotective effects of adenosine following cardiac arrest and resuscitation. Although adenosine did not prevent eventual CA1 neuronal loss in the long term, it did delay neuronal loss and promoted long-term survival. Thus, adenosine or specific agonists of adenosine receptors should be evaluated as adjuncts to broaden the window of opportunity in the treatment of the reperfusion injury following cardiac arrest and resuscitation.
- Published
- 2006
- Full Text
- View/download PDF
45. Neuronal migration is transiently delayed by prenatal exposure to intermittent hypoxia.
- Author
-
Zechel JL, Gamboa JL, Peterson AG, Puchowicz MA, Selman WR, and Lust WD
- Subjects
- Actins metabolism, Animals, Animals, Newborn, Body Weight, Catalase metabolism, Female, Glutathione Peroxidase metabolism, Immunoblotting, Immunoprecipitation, Maternal Exposure, Nicotine administration & dosage, Nicotine toxicity, Pregnancy, Pregnancy, Animal, Proto-Oncogene Proteins c-abl metabolism, Rats, Rats, Sprague-Dawley, Cell Movement physiology, Hypoxia metabolism, Neurons physiology, Prenatal Exposure Delayed Effects
- Abstract
Background: Neonatal neurodevelopment is influenced by a variety of external factors, although the mechanisms responsible are poorly understood. Prenatal hypoxia, from physiological or chemical sources, can have no discernible effect, or can result in a broad spectrum of abnormalities., Methods: To mimic some of the maternal effects of smoking, we developed a model that investigates the effects of intermittent hypoxia (IH), with or without concurrent nicotine in timed pregnant Sprague-Dawley rats., Results: We found no significant differences between litter sizes or birthweight of pups from any treatment group, but animals exposed to IH (with or without nicotine) showed long term diminished body weights. Animals subjected to IH consistently showed a transient delay in neuronal migration early in the postpartum period, which was amplified by concurrent nicotine administration. We observed increased c-Abl protein levels in animals from the IH treatment groups. Multiple proteins involved in the intricate control of neuronal migration were also altered in response to this treatment, primarily the downstream targets of c-Abl: Cdk5, p25, and the cytoskeletal elements neurofilament H and F-actin and catalase. Catalase activity and protein levels, already elevated in response to IH, were further amplified by simultaneous nicotine exposure., Conclusions: This new model provides a novel system for investigating the effects of low grade IH in the developing brain and suggests that concurrent nicotine further aggravates many of the deleterious effects of IH., (Copyright 2005 Wiley-Liss, Inc)
- Published
- 2005
- Full Text
- View/download PDF
46. Computational study on use of single-point analysis method for quantitating local cerebral blood flow in mice.
- Author
-
Puchowicz MA, Radhakrishnan K, Xu K, Magness DL, and LaManna JC
- Subjects
- Algorithms, Animals, Antipyrine analogs & derivatives, Autoradiography methods, Autoradiography statistics & numerical data, Carbon Radioisotopes, Male, Mice, Mice, Inbred C57BL, Models, Biological, Regional Blood Flow, Sensitivity and Specificity, Cerebrovascular Circulation
- Abstract
The benefits of a mouse model are efficiency and availability of transgenics/ knockouts. Quantitation of cerebral blood in small animals is difficult because the cannulation procedure may introduce errors. The [14C]-iodoantipyrine autoradiography (IAP) method requires both the tissue concentration and the time course of arterial concentration of the [14C] radioactive tracer. A single point-analysis technique was evaluated for measuring blood flow in mice (30 g +/- 0.3 g; n = 11) by using computational models of sensitivity analysis, which quantitates relationships between the predictions of a model and its parameters. Using [14C]-IAP in conjunction with mathematical algorithms and assumed arterial concentration-versus-time profiles, cortical blood flow was deduced from single-point measurements of the arterial tracer concentration. The data showed the arterial concentration profile that produced the most realistic blood flows (1.6 +/- 0.4; mean +/- SD, ml/g/min) was a profile with a ramp time of 30 sec followed by a constant value over the remaining time period of 30 sec. Sensitivity analysis showed that the total experimental time period was a more important parameter than the lag period and the ramp period. Thus, it appears that the accuracy of the assumption of linearly increasing arterial concentration depends on the experimental time period and the final arterial [14C]-iodoantipyrine concentration.
- Published
- 2005
- Full Text
- View/download PDF
47. Adaptation to chronic hypoxia during diet-induced ketosis.
- Author
-
Puchowicz MA, Emancipator DS, Xu K, Magness DL, Ndubuizu OI, Lust WD, and LaManna JC
- Subjects
- Adaptation, Physiological, Animals, Chronic Disease, Diet, Glucose metabolism, Glycolysis, Ketosis etiology, Lactic Acid metabolism, Male, Rats, Rats, Wistar, Hypoxia, Brain complications, Hypoxia, Brain metabolism, Ketosis complications
- Abstract
It is recognized that brain oxygen deprivation results in increased glycolysis and lactate accumulation. Moreover, glucose metabolism is altered during starvation or diet, resulting in increased plasma ketones (acetoacetate + beta-hydroxybutyrate; BHB). We investigated glucose and lactate adaptation to hypoxia in concurrence with diet-induced ketosis. Male Wistar rats were fed standard (STD), ketogenic (high fat; KG), or carbohydrate-rich (low fat; CHO) diets for 3 wks and then exposed to hypobaric (0.5 ATM) or normobaric atmosphere for 3 wks while on their diets. Lactate, ketones, and glucose concentrations were measured in plasma (mM) and brain tissue (mmol/g). Plasma and tissue ketone levels were elevated up to 12-fold in the KG fed groups compared with other groups (STD and CHO), with the hypoxic KG group reaching the highest levels (2.6 +/- 1.3 mM and 0.3 +/- 0.1 mmol/g; mean +/- SD). Tissue lactate levels in the hypoxic ketotic rats (4.7 +/- 1.3 mM) were comparable with normoxic STD (5.0 +/- 0.7 mM) and significantly lower (ANOVA P < .05) than the hypoxic STD rats (6.1 +/- 1.0 mM). These data indicate that adaptation to hypoxia did not interfere with ketosis, and that ketosis during hypoxia may lower lactate levels in brain, suggesting decreased glycolysis or increased glucose disposal.
- Published
- 2005
- Full Text
- View/download PDF
48. Renormalization of regional brain blood flow during prolonged mild hypoxic exposure in rats.
- Author
-
Xu K, Puchowicz MA, and LaManna JC
- Subjects
- Analysis of Variance, Animals, Blood Pressure physiology, Brain physiopathology, Hematocrit methods, Male, Oxygen metabolism, Rats, Rats, Wistar, Time Factors, Brain blood supply, Cerebrovascular Circulation physiology, Hyperoxia physiopathology, Regional Blood Flow physiology
- Abstract
In this study, we measured regional brain blood flow (BF) in rats during hypoxic exposure. Our data show the hypoxia-induced brain blood flow increase returned to baseline within 1 week as blood hemoglobin increased. Because this return to baseline occurred before capillary angiogenesis, this result suggests that the mechanism for brain blood flow renormalization during prolonged hypoxia exposure is more likely related to the increased systemic arterial oxygen carrying capacity than to local tissue hypoxia that persists for at least 2 weeks.
- Published
- 2004
- Full Text
- View/download PDF
49. Oxidative phosphorylation analysis: assessing the integrated functional activity of human skeletal muscle mitochondria--case studies.
- Author
-
Puchowicz MA, Varnes ME, Cohen BH, Friedman NR, Kerr DS, and Hoppel CL
- Abstract
Oxidative phosphorylation analysis, performed on freshly-isolated mitochondria, assesses the integrated function of the electron transport chain (ETC) coupled to ATP synthesis, membrane transport, dehydrogenase activities, and the structural integrity of the mitochondria. In this review, a case study approach is employed to highlight detection of defects in the adenine nucleotide translocator, the pyruvate dehydrogenase complex, fumarase, coenzyme Q function, fatty acid metabolism, and mitochondrial membrane integrity. Our approach uses the substrates glutamate, pyruvate, 2-ketoglutarate (coupled with malonate), malate, and fatty acid substrates (palmitoylcarnitine, octanoylcarnitine, palmitoyl-CoA (with carnitine), octanoyl-CoA (with carnitine), octanoate and acetylcarnitine) in addition to succinate, durohydroquinone and TMPD/ascorbate to uncover metabolic defects that would not be apparent from ETC assays performed on detergent-solubilized mitochondria.
- Published
- 2004
- Full Text
- View/download PDF
50. Comparison of glucose influx and blood flow in retina and brain of diabetic rats.
- Author
-
Puchowicz MA, Xu K, Magness D, Miller C, Lust WD, Kern TS, and LaManna JC
- Subjects
- Animals, Blood Glucose analysis, Blood-Brain Barrier metabolism, Cerebral Cortex metabolism, Diabetes Mellitus, Experimental metabolism, Male, Rats, Rats, Wistar, Retinal Vessels metabolism, Retinal Vessels physiology, Blood-Retinal Barrier metabolism, Cerebral Cortex blood supply, Cerebrovascular Circulation physiology, Diabetes Mellitus, Experimental physiopathology, Glucose metabolism, Retina metabolism
- Abstract
Diabetes is associated with extensive microvascular pathology and decreased expression of the glucose transporter (GLUT-1) in retina, but not brain. To explore the basis of these differences, the authors simultaneously measured glucose influx (micromol x g(-1) x min(-1)) and blood flow (mL x g(-1) x min(-1)) in retina and brain cortex of nondiabetic control rats (normoglycemic and acute-hyperglycemic) and in rats with streptozotocin-induced diabetes (with or without aminoguanidine (AMG) treatment) using a single-pass, dual-label indicator method. In addition, tissue glucose and adenosine triphosphate (nmol/mg dry weight) levels were measured. Glucose influx in retina exceeded that of cortex by about threefold for both the nondiabetic and diabetic groups. In contrast, blood flow in retina was significantly lower than in cortex by about threefold for each group. Retinal and cortical glucose influx in the diabetic rats was lower than in the nondiabetic acute-hyperglycemic group, but not in the normoglycemic group. Blood flow in these tissues remained relatively unchanged with glycemic conditions. The glucose levels in the diabetic retina (aminoguanidine untreated and aminoguanidine treated) were fourfold to sixfold greater than the nondiabetic retina. The cortical glucose levels remained unchanged in all groups. These data suggest that the accumulation of glucose in the diabetic retina cannot be explained by increased endothelial-glucose uptake or increased vascular membrane permeability.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.