Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Mol Cell Cardiol ; 158: 115-127, 2021 09.
Article in English | MEDLINE | ID: mdl-34081952

ABSTRACT

RATIONALE: The nutrient sensing mechanistic target of rapamycin complex 1 (mTORC1) and its primary inhibitor, tuberin (TSC2), are cues for the development of cardiac hypertrophy. The phenotype of mTORC1 induced hypertrophy is unknown. OBJECTIVE: To examine the impact of sustained mTORC1 activation on metabolism, function, and structure of the adult heart. METHODS AND RESULTS: We developed a mouse model of inducible, cardiac-specific sustained mTORC1 activation (mTORC1iSA) through deletion of Tsc2. Prior to hypertrophy, rates of glucose uptake and oxidation, as well as protein and enzymatic activity of glucose 6-phosphate isomerase (GPI) were decreased, while intracellular levels of glucose 6-phosphate (G6P) were increased. Subsequently, hypertrophy developed. Transcript levels of the fetal gene program and pathways of exercise-induced hypertrophy increased, while hypertrophy did not progress to heart failure. We therefore examined the hearts of wild-type mice subjected to voluntary physical activity and observed early changes in GPI, followed by hypertrophy. Rapamycin prevented these changes in both models. CONCLUSION: Activation of mTORC1 in the adult heart triggers the development of a non-specific form of hypertrophy which is preceded by changes in cardiac glucose metabolism.


Subject(s)
Cardiomegaly/metabolism , Gene Knockdown Techniques/methods , Glucose/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Signal Transduction/genetics , Animals , Cardiomegaly/diet therapy , Cardiomegaly/genetics , Cardiomegaly/prevention & control , Cells, Cultured , Diet/methods , Disease Models, Animal , Enzyme Activation/genetics , Glucose-6-Phosphatase/metabolism , Isomerases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Phosphorylation/genetics , Sirolimus/administration & dosage , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism
2.
Urol Ann ; 8(2): 245-8, 2016.
Article in English | MEDLINE | ID: mdl-27141204

ABSTRACT

Unicentric Castleman disease (UCD) is a relatively rare lymphoproliferative disease, which commonly presents as a mediastinal mass and less frequently involves abdomen, pelvis, and retroperitoneum. We report a case of a 64-year-old man with newly diagnosed low-volume, Gleason 3 + 3 = 6 prostate adenocarcinoma, who in considering active surveillance versus treatment was found to have a left perivesical and iliac chain lymphadenopathy concerning for potential metastatic involvement. He underwent magnetic resonance imaging with ferumoxytol to assist in the diagnostic evaluation to better characterize his lymphadenopathy. Subsequently, he underwent robotic-assisted laparoscopic bilateral pelvic lymph node dissection and resection of left perivesical mass exhibiting hyaline vascular variant of UCD.

3.
FASEB J ; 27(10): 3966-78, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23825227

ABSTRACT

Diabetic patients with acute myocardial infarction are more likely to die than nondiabetic patients. In the present study we examined the effect of insulin resistance on myocardial ischemia tolerance. Hearts of rats, rendered insulin resistant by high-sucrose feeding, were subjected to ischemia/reperfusion ex vivo. Cardiac power of control hearts from chow-fed rats recovered to 93%, while insulin-resistant hearts recovered only to 80% (P<0.001 vs. control). Unexpectedly, impaired contractile recovery did not result from an impairment of glucose oxidation (576±36 vs. 593±42 nmol/min/g dry weight; not significant), but from a failure to increase and to sustain oxidation of the long-chain fatty acid oleate on reperfusion (1878±56 vs. 2070±67 nmol/min/g dry weight; P<0.05). This phenomenon was due to a reduced ability to transport oleate into mitochondria and associated with a 38-58% decrease in the mitochondrial uncoupling protein 3 (UCP3) levels. Contractile function was rescued by replacing oleate with a medium-chain fatty acid or by restoring UCP3 levels with 24 h of food withdrawal. Lastly, the knockdown of UCP3 in rat L6 myocytes also decreased oleate oxidation by 13-18% following ischemia. Together the results expose UCP3 as a critical regulator of long-chain fatty acid oxidation in the stressed heart postischemia and identify octanoate as an intervention by which myocardial metabolism can be manipulated to improve function of the insulin-resistant heart.


Subject(s)
Fatty Acids/chemistry , Fatty Acids/metabolism , Insulin Resistance/physiology , Reperfusion Injury/metabolism , Animal Feed , Animals , Caprylates , Diet , Dietary Carbohydrates , Gene Expression Regulation , Glucose/metabolism , Insulin/metabolism , Ion Channels/genetics , Ion Channels/metabolism , Male , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidation-Reduction , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Sucrose , Uncoupling Protein 3
4.
J Vasc Surg ; 58(3): 766-75.e12, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23755976

ABSTRACT

OBJECTIVE: The present study is designed to understand the contribution of peripheral vascular disease and peripheral neuropathy to the wound-healing impairment associated with diabetes. Using a rabbit model of diabetic neuroischemic wound healing, we investigated rate of healing, leukocyte infiltration, and expression of cytokines, interleukin-8 and interleukin-6, and neuropeptides, substance P, and neuropeptide Y. METHODS: Diabetes was induced in New Zealand White rabbits by administering alloxan while control rabbits received saline. Ten days later, animals in both groups underwent surgery. One ear served as a sham, and the other was made ischemic (ligation of central+rostral arteries) or neuroischemic (ischemia+ resection of central+rostral nerves). Four 6-mm punch biopsy wounds were created in both ears and wound healing was followed for 10 days using computerized planimetry. RESULTS: Nondiabetic sham and ischemic wounds healed significantly more rapidly than diabetic sham and ischemic wounds. Healing was slowest in neuroischemic wounds, irrespective of diabetic status. A high M1/M2 macrophage ratio and a high proinflammatory cytokine expression, both indicators of chronic proinflammatory state, and low neuropeptide expression were seen in preinjury diabetic skin. Postinjury, in diabetic wounds, the M1/M2 ratio remained high, the reactive increase in cytokine expression was low, and neuropeptide expression was further decreased in neuroischemic wounds. CONCLUSIONS: This rabbit model illustrates how a combination of a high M1/M2 ratio, a failure to mount postinjury cytokine response as well as a diminished neuropeptide expression, contribute to wound-healing impairment in diabetes. The addition of neuropathy to ischemia leads to equivalently severe impaired wound-healing irrespective of diabetes status, suggesting that in the presence of ischemia, loss of neuropeptide function contributes to the impaired healing associated with diabetes.


Subject(s)
Cytokines/metabolism , Diabetes Mellitus, Experimental/complications , Diabetic Angiopathies/etiology , Diabetic Neuropathies/etiology , Inflammation Mediators/metabolism , Ischemia/etiology , Neuropeptides/metabolism , Skin Ulcer/etiology , Skin , Wound Healing , Animals , Cytokines/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Diabetic Angiopathies/genetics , Diabetic Angiopathies/immunology , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Diabetic Neuropathies/genetics , Diabetic Neuropathies/immunology , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/pathology , Down-Regulation , Ischemia/genetics , Ischemia/immunology , Ischemia/metabolism , Ischemia/pathology , Macrophages/immunology , Neuropeptides/genetics , Rabbits , Skin/immunology , Skin/metabolism , Skin/pathology , Skin Ulcer/genetics , Skin Ulcer/immunology , Skin Ulcer/metabolism , Skin Ulcer/pathology , Time Factors , Up-Regulation
5.
J Am Heart Assoc ; 2(3): e004796, 2013 May 17.
Article in English | MEDLINE | ID: mdl-23686371

ABSTRACT

BACKGROUND: Changes in energy substrate metabolism are first responders to hemodynamic stress in the heart. We have previously shown that hexose-6-phosphate levels regulate mammalian target of rapamycin (mTOR) activation in response to insulin. We now tested the hypothesis that inotropic stimulation and increased afterload also regulate mTOR activation via glucose 6-phosphate (G6P) accumulation. METHODS AND RESULTS: We subjected the working rat heart ex vivo to a high workload in the presence of different energy-providing substrates including glucose, glucose analogues, and noncarbohydrate substrates. We observed an association between G6P accumulation, mTOR activation, endoplasmic reticulum (ER) stress, and impaired contractile function, all of which were prevented by pretreating animals with rapamycin (mTOR inhibition) or metformin (AMPK activation). The histone deacetylase inhibitor 4-phenylbutyrate, which relieves ER stress, also improved contractile function. In contrast, adding the glucose analogue 2-deoxy-d-glucose, which is phosphorylated but not further metabolized, to the perfusate resulted in mTOR activation and contractile dysfunction. Next we tested our hypothesis in vivo by transverse aortic constriction in mice. Using a micro-PET system, we observed enhanced glucose tracer analog uptake and contractile dysfunction preceding dilatation of the left ventricle. In contrast, in hearts overexpressing SERCA2a, ER stress was reduced and contractile function was preserved with hypertrophy. Finally, we examined failing human hearts and found that mechanical unloading decreased G6P levels and ER stress markers. CONCLUSIONS: We propose that glucose metabolic changes precede and regulate functional (and possibly also structural) remodeling of the heart. We implicate a critical role for G6P in load-induced mTOR activation and ER stress.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Glucose/physiology , Heart/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Humans , In Vitro Techniques , Male , Rats , Rats, Sprague-Dawley , Signal Transduction
6.
PLoS One ; 7(6): e39123, 2012.
Article in English | MEDLINE | ID: mdl-22720046

ABSTRACT

Vein graft failure occurs between 1 and 6 months after implantation due to obstructive intimal hyperplasia, related in part to implantation injury. The cell-specific and temporal response of the transcriptome to vein graft implantation injury was determined by transcriptional profiling of laser capture microdissected endothelial cells (EC) and medial smooth muscle cells (SMC) from canine vein grafts, 2 hours (H) to 30 days (D) following surgery. Our results demonstrate a robust genomic response beginning at 2 H, peaking at 12-24 H, declining by 7 D, and resolving by 30 D. Gene ontology and pathway analyses of differentially expressed genes indicated that implantation injury affects inflammatory and immune responses, apoptosis, mitosis, and extracellular matrix reorganization in both cell types. Through backpropagation an integrated network was built, starting with genes differentially expressed at 30 D, followed by adding upstream interactive genes from each prior time-point. This identified significant enrichment of IL-6, IL-8, NF-κB, dendritic cell maturation, glucocorticoid receptor, and Triggering Receptor Expressed on Myeloid Cells (TREM-1) signaling, as well as PPARα activation pathways in graft EC and SMC. Interactive network-based analyses identified IL-6, IL-8, IL-1α, and Insulin Receptor (INSR) as focus hub genes within these pathways. Real-time PCR was used for the validation of two of these genes: IL-6 and IL-8, in addition to Collagen 11A1 (COL11A1), a cornerstone of the backpropagation. In conclusion, these results establish causality relationships clarifying the pathogenesis of vein graft implantation injury, and identifying novel targets for its prevention.


Subject(s)
Transcriptome , Veins/transplantation , Animals , Dogs , Quality Control
7.
FASEB J ; 26(8): 3118-26, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22611083

ABSTRACT

Insulin resistance is a prominent feature in heart failure, while hyperglycemia impairs cardiac contraction. We propose that decreased insulin-mediated glucose uptake by the heart preserves cardiac function in response to metabolic and hemodynamic stress. To test this hypothesis, we fed rats a high-sucrose diet (HSD). Energy substrate metabolism and cardiac work were determined ex vivo in a sequential protocol simulating metabolic and hemodynamic stress. Compared to chow-fed, control rats, HSD impaired myocardial insulin responsiveness and induced profound metabolic changes in the heart, characterized by reduced rates of glucose uptake (7.91 ± 0.30 vs. 10.73 ± 0.67 µmol/min/g dry weight; P<0.001) but increased rates of glucose oxidation (2.38 ± 0.17 vs. 1.50 ± 0.15 µmol/min/g dry weight; P<0.001) and oleate oxidation (2.29 ± 0.11 vs. 1.96 ± 0.12 µmol/min/g dry weight; P<0.05). Tight coupling of glucose uptake and oxidation and improved cardiac efficiency were associated with a reduction in glucose 6-phosphate and oleoyl-CoA levels, as well as a reduction in the content of uncoupling protein 3. Our results suggest that insulin resistance lessens fuel toxicity in the stressed heart. This calls for a new exploration of the mechanisms regulating substrate uptake and oxidation in the insulin-resistant heart.


Subject(s)
Heart/physiology , Insulin Resistance/physiology , Myocardium/metabolism , Stress, Physiological/physiology , Animals , Dietary Sucrose/administration & dosage , Dietary Sucrose/pharmacology , Down-Regulation , Glucose/metabolism , In Vitro Techniques , Insulin/physiology , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Myocardial Contraction/drug effects , Oleic Acid/metabolism , Perfusion , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Uncoupling Protein 3
8.
J Vasc Surg ; 53(6): 1654-60.e2, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21609799

ABSTRACT

BACKGROUND: Impaired wound healing is a major complication associated with diabetes, involving a dysregulation and impairments in the inflammatory and angiogenic phases of wound healing. Here, we examine the effects of the neuropeptides substance P (SP) and neuropeptide Y (NPY) on dermal microvascular endothelial cell (DMVEC) angiogenesis and interleukin-8 (IL-8) expression, a known effector of the neuropeptide pathways in normal and hyperglycemic conditions in vitro. METHODS: DMVECs are treated with one of four glucose concentrations: 1) 5 mM glucose; 2) 10 mM glucose; 3) 30 mM glucose; or 4) 30 mM mannitol and cotreated with 100 nM NPY, 100 nM SP, or 10 ng/mL IL-8. Angiogenesis is assessed with proliferation and tube formation assays. IL-8 mRNA and protein expression are evaluated at days 1 and 7. RESULTS: As compared with noromoglycemia (5 mM glucose), hyperglycemia (30 mM glucose) decreases DMVEC proliferation and tube formation by 39% and 42%, respectively. SP cotreatment restores DMVEC proliferation (211%) and tube formation (152%), and decreases IL-8 expression (34%) in DMVECs exposed to hyperglycemic conditions. These effects are not observed with NPY. However, IL-8 treatment by itself does not affect proliferation or tube formation, suggesting that the effect of SP on DMVEC angiogenesis is unlikely through changes in IL-8 expression. CONCLUSION: Hyperglycemic conditions impair DMVEC proliferation and tube formation. SP mitigates the effect of hyperglycemia on DMVECs by increasing DMVEC proliferation and tube formation. These findings are not likely to be related to a dysregulation of IL-8 due to the lack of effects of hyperglycemia on IL-8 expression and the lack of effect of IL-8 on DMVEC proliferation and tube formation. The effect of SP on DMVECs makes SP a promising potential target for therapy in impaired wound healing in diabetes, but the exact mechanism remains unknown.


Subject(s)
Endothelium, Vascular/metabolism , Hyperglycemia/metabolism , Interleukin-8/biosynthesis , Neovascularization, Physiologic/physiology , Neuropeptides/metabolism , Wound Healing/physiology , Cell Proliferation , Cells, Cultured , Dermis/blood supply , Dermis/metabolism , Endothelial Cells/physiology , Endothelium, Vascular/physiology , Endothelium, Vascular/physiopathology , Humans , Hyperglycemia/physiopathology , Microvessels/metabolism , Neuropeptide Y/metabolism , Substance P/metabolism
9.
J Surg Res ; 167(2): 336-42, 2011 May 15.
Article in English | MEDLINE | ID: mdl-20070982

ABSTRACT

The interaction between neuropeptides and cytokines and its role in cutaneous wound healing is becoming evident. The goal of the present study is to investigate the impact of diabetes on peripheral cytokine and neuropeptide expression and its role in diabetic wound healing. To achieve this goal, the effect of diabetes on wound healing, along with the role of inflammatory cytokines such as interleukin-6 (IL-6) and interleukin-8 (IL-8) secreted in the wound microenvironment, and neuropeptides such as substance P (SP) and neuropeptide Y (NPY), secreted from peripheral nerves is monitored in non-diabetic and diabetic rabbits. Rabbits in the diabetic group received alloxan monohydrate (100mg/kg i.v.). Ten days after diabetic induction, four full thickness circular wounds were created in both ears using a 6mm punch biopsy. Wound healing was monitored over 10 d and gene expression of cytokines and neuropeptides was assessed in the wounds. Compared with the non-diabetic rabbits, wounds of diabetic rabbits heal significantly slower. Diabetic rabbits show significantly increased baseline gene expression of IL-6 and IL-8, their receptors, CXCR1, CXCR2, GP-130, and a decrease of prepro tachykinin-A (PP-TA), the precursor of SP, whereas the expression of prepro-NPY (PP-NPY), the precursor of NPY is not different. Similarly, baseline protein expression of CXCR1 is higher in diabetic rabbit skin. Post-injury, the increase over baseline gene expression of IL-6, IL-8, CXCR1, CXCR2, and GP-130 is significantly less in diabetic wounds compared with non-diabetic wounds. Although there is no difference in PP-TA gene expression between non-diabetic and diabetic rabbits post-injury, the gene expression of PP-NPY is reduced in diabetic rabbits. In conclusion, diabetes causes dysregulation in the neuropeptide expression in the skin along with a suppressed focused inflammatory response to injury. This suggests that the chronic inflammation in the skin of diabetic rabbits inhibits the acute inflammation much needed for wound healing.


Subject(s)
Cytokines/metabolism , Diabetes Mellitus, Experimental/metabolism , Inflammation/metabolism , Neuropeptides/metabolism , Wound Healing/physiology , Alloxan , Animals , Disease Models, Animal , Interleukin-6/metabolism , Interleukin-8/metabolism , Neuropeptide Y/metabolism , Rabbits , Substance P/metabolism
10.
Am J Physiol Heart Circ Physiol ; 295(1): H256-65, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18487439

ABSTRACT

The cardiac-enriched isoform of acetyl-CoA carboxylase (ACC2) is a key regulator of mitochondrial fatty acid (FA) uptake via carnitine palmitoyltransferase 1 (CPT1). To test the hypothesis that oxidative metabolism is upregulated in hearts from animals lacking ACC2 (employing a transgenic Acc2-mutant mouse), we assessed cardiac function in vivo and determined rates of myocardial substrate oxidation ex vivo. When examined by echocardiography, there was no difference in systolic function, but left ventricular mass of the Acc2-mutant (MUT) mouse was significantly reduced ( approximately 25%) compared with wild-types (WT). Reduced activation of the mammalian target of rapamycin (mTOR) and its downstream target p70S6K was found in MUT hearts. Exogenous oxidation rates of oleate were increased approximately 22%, and, unexpectedly, exogenous glucose oxidation rates were also increased in MUT hearts. Using a hyperinsulinemic-euglycemic clamp, we found that glucose uptake in MUT hearts was increased by approximately 83%. Myocardial triglyceride levels were significantly reduced in MUT vs. WT while glycogen content was the same. In parallel, transcript levels of PPARalpha and its target genes, pyruvate dehydrogenase kinase-4 (PDK-4), malonyl-CoA decarboxylase (MCD), and mCPT1, were downregulated in MUT mice. In summary, we report that 1) Acc2-mutant hearts exhibit a marked preference for the oxidation of both glucose and FAs coupled with greater utilization of endogenous fuel substrates (triglycerides), 2) attenuated mTOR signaling may result in reduced heart sizes observed in Acc2-mutant mice, and 3) Acc2-mutant hearts displayed normal functional parameters despite a significant decrease in size.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Energy Metabolism , Mutation , Myocardium/enzymology , Acetyl-CoA Carboxylase/genetics , Animals , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Down-Regulation , Glucose/metabolism , Glucose Clamp Technique , Glycogen/metabolism , Heart Ventricles/diagnostic imaging , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/pathology , Oleic Acid/metabolism , Organ Size , Oxidation-Reduction , PPAR gamma/genetics , PPAR gamma/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases , Time Factors , Triglycerides/metabolism , Ultrasonography
11.
Cardiovasc Res ; 76(1): 71-80, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17553476

ABSTRACT

OBJECTIVE: Insulin regulates both glucose uptake and postnatal cardiac growth. The anabolic effects of insulin are mediated by the mammalian target of rapamycin (mTOR), an evolutionarily conserved kinase which is also a convergence point between nutrient sensing and cell growth. We postulated that mTOR signalling in the heart requires the metabolism of glucose. METHODS: We interrogated the insulin-mediated mTOR signalling pathway in response to different metabolic interventions regulating substrate metabolism in the isolated working rat heart and in isolated cardiomyocytes. RESULTS: Although insulin enhanced Akt activity, phosphorylation of mTOR and its downstream targets (p70S6K and 4EBP1) required the addition of glucose. Glucose-dependent p70S6K phosphorylation was independent of the hexosamine biosynthetic pathway, the AMP kinase pathway, and the pentose phosphate pathway. However, inhibition of glycolysis downstream of hexokinase markedly enhanced p70S6K phosphorylation. Furthermore, 2-deoxyglucose activated p70S6K suggesting that phosphorylation of glucose is required for carbohydrate-mediated mTOR signalling in the heart. Lastly, we also found enhanced p70S6K phosphorylation in the hearts of diabetic rats. CONCLUSION: Phosphorylation of glucose is necessary for insulin-dependent mTOR activity in the heart, suggesting a link between intermediary metabolism and cardiac growth.


Subject(s)
Glucose/metabolism , Insulin/metabolism , Myocytes, Cardiac/metabolism , Protein Kinases/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Newborn , Blotting, Western/methods , Cell Cycle Proteins , Cells, Cultured , Glycolysis , Hexosamines/biosynthesis , Male , Pentosephosphates/metabolism , Phosphoproteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases
12.
Gene Regul Syst Bio ; 1: 251-62, 2007 Nov 05.
Article in English | MEDLINE | ID: mdl-19936093

ABSTRACT

BACKGROUND: During pressure overload-induced hypertrophy, unloading-induced atrophy, and diabetes mellitus, the heart induces 'fetal' genes (e.g. myosin heavy chain beta; mhc beta). HYPOTHESIS: We propose that altered glucose homeostasis within the cardiomyocyte acts as a central mechanism for the regulation of gene expression in response to environmental stresses. The evidence is as follows. METHODS AND RESULTS: Forced glucose uptake both ex vivo and in vivo results in mhc isoform switching. Restricting dietary glucose prevents mhc isoform switching in hearts of both GLUT1-Tg mice and rats subjected to pressure overload-induced hypertrophy. Thus, glucose availability correlates with mhc isoform switching under all conditions investigated. A potential mechanism by which glucose affects gene expression is through O-linked glycosylation of specific transcription factors. Glutamine:fructose-6-phosphate amidotransferase (GFAT) catalyzes the flux generating step in UDP-N-acetylglucosamine biosynthesis, the rate determining metabolite in protein glycosylation. Ascending aortic constriction increased intracellular levels of UDP-N-acetylglucosamine, and the expression of gfat2, but not gfat1, in the rat heart. CONCLUSIONS: Collectively, the results strongly suggest glucose-regulated gene expression in the heart, and the involvement of glucose metabolites in isoform switching of sarcomeric proteins characteristic for the fetal gene program.

13.
J Proteome Res ; 5(1): 177-82, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16396509

ABSTRACT

The discovery of novel serological biomarkers is critical for improving disease diagnosis and monitoring treatment response. Proteomic analysis of model systems, such as isolated cells in culture and patient plasma and serum, represents the current state-of-the-art. Here, we coupled proteomics with isolated organ perfusion, which allows a disease state to be studied in a physiologic, yet controlled, environment. Potential markers specific to the disease or to changes in the surrounding tissue may be discovered. The effectiveness of this model was evaluated using proteomic analysis of effluent fractions collected from isolated beating rat hearts during reperfusion after brief episodes of ischemia. The detection of clinical markers for myocardial ischemia in this effluent was robust and analytically straightforward, validating the potential of isolated organ perfusion in diagnostic protein discovery.


Subject(s)
Myocardial Ischemia/diagnosis , Myocardium/chemistry , Perfusion , Proteins/analysis , Proteome/analysis , Proteomics , Animals , Biomarkers/analysis , In Vitro Techniques , Male , Rats , Rats, Wistar
14.
Am J Physiol Endocrinol Metab ; 289(2): E328-36, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15797988

ABSTRACT

It is suggested that insulin resistance and metabolic maladaptation of the heart are causes of contractile dysfunction. We tested the hypothesis whether systemic PPARgamma activation, by changing the metabolic profile in a model of insulin resistance and type 2 diabetes (the ZDF rat) in vivo, improves contractile function of the heart in vitro. Male Zucker diabetic fatty (ZDF) and Zucker lean (ZL) rats, at 53-56 days of age, were treated with either GI-262570 (a nonthiazolidinedione PPARgamma agonist; A) or vehicle (V) for 1 wk. Agonist treatment resulted in correction of hyperglycemia and dyslipidemia, as well as in reduced hyperinsulinemia. The accumulation of triacylglycerols in the myocardium, characteristic of the ZDF rat, disappeared with treatment. Cardiac power and rates of glucose oxidation in the isolated working heart were significantly reduced in ZDF-V rats, but both parameters increased to nondiabetic levels with agonist treatment. In ZDF-V hearts, transcript levels of PPARalpha-regulated genes and of myosin heavy chain-beta were upregulated, whereas GLUT4 was downregulated compared with ZL. Agonist treatment of ZDF rats reduced PPARalpha-regulated genes and increased transcripts of GLUT4 and GLUT1. In conclusion, by changing the metabolic profile, reducing myocardial lipid accumulation, and promoting the downregulation of PPARalpha-regulated genes, PPARgamma activation leads to an increased capacity of the myocardium to oxidize glucose and to a tighter coupling of oxidative metabolism and contraction in the setting of insulin resistance and type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Myocardial Contraction/physiology , Myocardium/metabolism , PPAR gamma/metabolism , Adaptation, Physiological , Animals , Disease Models, Animal , Energy Metabolism/drug effects , Energy Metabolism/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , In Vitro Techniques , Insulin Resistance/physiology , Male , Myocardial Contraction/drug effects , Oxazoles/pharmacology , PPAR gamma/agonists , Rats , Rats, Inbred Strains , Rats, Zucker , Signal Transduction/drug effects , Signal Transduction/physiology , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Triglycerides/metabolism , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
15.
Am J Physiol Endocrinol Metab ; 285(6): E1312-6, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12944323

ABSTRACT

To what extent does glutamine turnover keep pace with oxidative metabolism in the rat heart? To address this question, the following groups of substrates were presented to the isolated, working rat heart: 1) glucose (5 mM), insulin (40 microU/ml), and [2-13C]acetate (5 mM; high workload, n = 5); 2) pyruvate (2.5 mM) and [2-13C]acetate (5 mM; normal workload, n = 5); or 3) propionate (1 mM) and [2-13C]acetate (2.5 mM; normal workload, n = 3). In a subset of these experiments, the exchange of glutamate and glutamine was quantified by separation with ion exchange chromatography and analysis by GC-MS. There was an apparent equilibration of mass isotopomers of glutamate and glutamine after 50 min of perfusion, although the extent of equilibration was not determined. The fractional enrichment in glutamine was 31% of the enrichment of glutamate with the three different perfusates. From high-resolution nuclear magnetic resonance spectra, we found a ratio of glutamine to glutamate content of 94.1, 53.4, and 96.9%, respectively, for each experimental group. In experiments for which l-[1-13C]glutamine (5 mM) was included in the perfusate of group 2, [1-13C]glutamine was detected in the heart, but transfer of 13C from glutamine to glutamate was not detected (n = 4). We conclude that, in the perfused working heart, production of glutamine by amidation of glutamate takes place and can be detected, whereas the reverse process, generation of glutamate from glutamine, remains undetected.


Subject(s)
Glutamic Acid/metabolism , Glutamine/metabolism , Heart/physiology , Myocardium/metabolism , Adaptation, Physiological , Animals , Cardiac Output/physiology , Glucose/metabolism , In Vitro Techniques , Insulin/metabolism , Male , Oxygen Consumption/physiology , Propionates/metabolism , Pyruvic Acid/metabolism , Rats , Rats, Sprague-Dawley , Sodium Acetate/metabolism , Substrate Cycling/physiology
16.
J Biol Chem ; 278(15): 13143-50, 2003 Apr 11.
Article in English | MEDLINE | ID: mdl-12522136

ABSTRACT

The addition of glutamine as a major nutrient to cultured neonatal rat cardiomyocytes produced an increase in myocyte size and the organization of actin into myofibrillar arrays. The cellular response was associated with increased abundance of the mRNAs encoding the contractile proteins, alpha-myosin heavy chain and cardiac alpha-actin, and the metabolic enzymes, muscle carnitine palmitoyl transferase I and muscle adenylosuccinate synthetase (ADSS1). Adss1 gene expression was induced approximately 5-fold in glutamine-treated rat neonatal cardiac myocytes. The induction was mediated through the protein kinase A and mammalian target of rapamycin signaling pathways and required a cyclic AMP response element associated with the promoter region of the Adss1 gene. These results highlight glutamine as a major nutrient regulator of cardiac gene expression and identify protein kinase A and mammalian target of rapamycin signaling pathways as mediators of the cardiomyocyte transcriptional response.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation/drug effects , Glutamine/pharmacology , Heart/physiology , Protein Kinases/metabolism , Transcription, Genetic/physiology , Adenylosuccinate Synthase/genetics , Angiotensin II/pharmacology , Animals , Cell Division/drug effects , Cells, Cultured , Heart/drug effects , Models, Biological , Myocardium/cytology , Myosin Heavy Chains/genetics , RNA, Messenger/genetics , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , TOR Serine-Threonine Kinases , Transcription, Genetic/drug effects
17.
Diabetes ; 51(8): 2587-95, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12145175

ABSTRACT

We investigated whether decreased responsiveness of the heart to physiological increases in fatty acid availability results in lipid accumulation and lipotoxic heart disease. Lean and obese Zucker rats were either fed ad libitum or fasted overnight. Fasting increased plasma nonesterified fatty acid levels in both lean and obese rats, although levels were greatest in obese rats regardless of nutritional status. Despite increased fatty acid availability, the mRNA transcript levels of peroxisome proliferator-activated receptor (PPAR)-alpha-regulated genes were similar in fed lean and fed obese rat hearts. Fasting increased expression of all PPAR-alpha -regulated genes in lean Zucker rat hearts, whereas, in obese Zucker rat hearts, muscle carnitine palmitoyltransferase and medium-chain acyl-CoA dehydrogenase were unaltered with fasting. Rates of oleate oxidation were similar for hearts from fed rats. However, fasting increased rates of oleate oxidation only in hearts from lean rats. Dramatic lipid deposition occurred within cardiomyocytes of obese, but not lean, Zucker rats upon fasting. Cardiac output was significantly depressed in hearts isolated from obese rats compared with lean rats, regardless of nutritional status. Fasting increased cardiac output in hearts of lean rats only. Thus, the heart's inability to increase fatty acid oxidation in proportion to increased fatty acid availability is associated with lipid accumulation and contractile dysfunction of the obese Zucker rat.


Subject(s)
Fatty Acids, Nonesterified/blood , Fatty Acids/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation , Muscle, Skeletal/physiopathology , Obesity/physiopathology , Acyl-CoA Dehydrogenase , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Animals , Carnitine O-Palmitoyltransferase , Carrier Proteins/genetics , Fasting , In Vitro Techniques , Ion Channels , Isoenzymes/genetics , Lipid Metabolism , Male , Mitochondrial Proteins , Muscle, Skeletal/enzymology , Myocardial Contraction , Myocardium/metabolism , Obesity/genetics , Obesity/metabolism , Oxidation-Reduction , Oxygen Consumption , Protein Kinases/genetics , Rats , Rats, Zucker , Receptors, Cytoplasmic and Nuclear/physiology , Thinness/genetics , Transcription Factors/physiology , Transcription, Genetic , Uncoupling Protein 3
18.
J Mol Cell Cardiol ; 34(2): 223-31, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11851361

ABSTRACT

The heart, like other organs, possesses an internal circadian clock. These clocks provide the selective advantage of anticipation, enabling the organ to prepare for a given stimulus, thereby optimizing the appropriate response. The heart in diabetes is associated with alterations in morphology, gene expression, metabolism and contractile performance. The present study investigated whether diabetes also alters the circadian clock in the heart. Insulin-dependent diabetes mellitus was induced in rats by treatment with streptozotocin (STZ; 65 mg/kg). STZ increased humoral (glucose and non-esterified fatty acids) and heart gene expression (myosin heavy chain beta, pyruvate dehydrogenase kinase 4 and uncoupling protein 3) markers of diabetes. The circadian patterns of gene expression of seven components of the mammalian clock (bmal1, clock, cry1, cry2, per1, per2 and per3), as well as three clock output genes (dbp, hlf and tef), were compared in hearts isolated from control and STZ-induced diabetic rats. All components of the clock investigated possessed circadian rhythms of gene expression. In the hearts isolated from STZ-induced diabetic rats, the phases of these circadian rhythms were altered (approximately 3 h early) compared to those observed for control hearts. The clock in the heart has therefore lost normal synchronization with its environment during diabetes. Whether this loss of synchronization plays a role in the development of contractile dysfunction of the heart in diabetes remains to be determined.


Subject(s)
Circadian Rhythm/physiology , Diabetes Mellitus, Experimental/physiopathology , Drosophila Proteins , Eye Proteins , Heart/physiology , Photoreceptor Cells, Invertebrate , Animals , Circadian Rhythm/genetics , Cryptochromes , Flavoproteins/physiology , Gene Expression Regulation/physiology , Male , Rats , Rats, Wistar , Receptors, G-Protein-Coupled
SELECTION OF CITATIONS
SEARCH DETAIL
...