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










Publication year range
3.
Toxicol Lett ; 284: 10-20, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29174818

ABSTRACT

Diabetes mellitus leads to oxidative stress and contractile dysfunction in the heart. Although several rationales have been speculated, the precise mechanism behind diabetic cardiomyopathy remains elusive. This study was designed to assess the role of inhibition of advanced glycation endproducts (AGE) in streptozotocin (STZ)-induced diabetic cardiac dysfunction. Cardiac contractile function was assessed in normal C57BL/6 and STZ (200mg/kg, single injection and maintained for 2 wks)-induced diabetic mice treated with or without the AGE inhibitor aminoguanidine (50mg/kg/d in drinking water) for 2 weeks using echocardiography and IonOptix MyoCam techniques. Diabetes compromised cardiac contractile function shown as reduced fractional shortening and ejection fraction, enlarged left ventricular end systolic/diastolic diameters, decreased peak shortening, maximal velocity of shortening/relengthening, prolonged shortening and relengthening duration as well as impaired intracellular Ca2+ homeostasis, the effects of which were alleviated or reversed by aminoguanidine treatment. Diabetes also inhibited autophagy, increased ER stress and phosphorylation of pro-hypertrophic signaling molecules Akt and mTOR, the effect of which was reversed by aminoguanidine. In vitro study revealed that methylglyoxal-derived AGE (MG-AGE) incubation in isolated cardiomyocytes promoted oxidation of sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA2a) and production of superoxide, the effects of which were negated by the autophagy inducer rapamycin, the ER stress chaperone TUDCA or the antioxidant N-acetylcysteine. Taken together, these data revealed that inhibition of AGE formation rescues against experimental diabetes-induced cardiac remodeling and contractile dysfunction possible through regulation of autophagy and ER stress.


Subject(s)
Autophagy/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetic Cardiomyopathies/prevention & control , Endoplasmic Reticulum Stress/drug effects , Glycation End Products, Advanced/antagonists & inhibitors , Guanidines/pharmacology , Animals , Calcium/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Echocardiography , Male , Mice, Inbred C57BL , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Streptozocin
4.
J Hepatol ; 62(3): 647-56, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25457208

ABSTRACT

BACKGROUND & AIMS: Mitochondrial aldehyde dehydrogenase (ALDH2) plays a critical role in the detoxification of the ethanol metabolite acetaldehyde. This study was designed to examine the impact of global ALDH2 overexpression on alcohol-induced hepatic steatosis. METHODS: Wild type Friend virus B (FVB) and ALDH2 transgenic mice were placed on a 4% alcohol or control diet for 12 weeks. Serum levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), bilirubin and cholesterol, hepatic triglyceride, steatosis, fat metabolism-related proteins, pro-inflammatory cytokines, glutathione (GSH), oxidized glutathione (GSSG), autophagy and autophagy signalling were examined. The role of autophagy was evaluated in alcohol dehydrogenase 1 (ADH1)-transfected human hepatocellular liver carcinoma cells (VA-13) treated with or without the autophagy inducer rapamycin and lysosomal inhibitors. RESULTS: Chronic alcohol intake led to elevated AST-, ALT-levels, bilirubin, AST/ALT ratio, cholesterol, hepatic triglycerides and hepatic fat deposition as evidenced by H&E and Oil Red O staining. Hepatic fat deposition was associated with disturbed levels of fat metabolism-related proteins (fatty acid synthase, SCD1), upregulated interleukin-6, TNF-α, cyclooxygenase, oxidative stress, and loss of autophagy, effects which were attenuated or ablated by the ALDH2 transgene. Moreover, ethanol (100 mM) and acetaldehyde (100 and 500 µM) increased levels of IL-6 and IFN-γ, and suppressed autophagy in VA-13 cells, effects which were markedly alleviated by rapamycin. In addition, lysosomal inhibitors mimicked ethanol-induced p62 accumulation with little additive effect with ethanol. Ethanol significantly suppressed LC3 conversion in the presence of lysosomal inhibitors. CONCLUSIONS: In summary, our results revealed that ALDH2 plays a beneficial role in ameliorating chronic alcohol intake-induced hepatic steatosis and inflammation through regulation of autophagy.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Autophagy/physiology , Liver Cirrhosis, Alcoholic/enzymology , Liver Cirrhosis, Experimental/enzymology , Acetaldehyde/metabolism , Alcoholism/complications , Alcoholism/metabolism , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase, Mitochondrial , Animals , Autophagy/drug effects , Cholesterol/blood , Cytokines/metabolism , Female , Hep G2 Cells , Humans , Lipid Metabolism , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Alcoholic/genetics , Liver Cirrhosis, Alcoholic/pathology , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/pathology , Lysosomes/drug effects , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Transgenic , Multiprotein Complexes/metabolism , Oxidative Stress , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Up-Regulation
5.
Biochim Biophys Acta ; 1852(2): 319-31, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24874076

ABSTRACT

Mitochondrial aldehyde dehydrogenase (ALDH2) is known to offer myocardial protection against stress conditions including ischemia-reperfusion injury, alcoholism and diabetes mellitus although the precise mechanism is unclear. This study was designed to evaluate the effect of ALDH2 on diabetes-induced myocardial injury with a focus on autophagy. Wild-type FVB and ALDH2 transgenic mice were challenged with streptozotozin (STZ, 200mg/kg, i.p.) for 3months to induce experimental diabetic cardiomyopathy. Diabetes triggered cardiac remodeling and contractile dysfunction as evidenced by cardiac hypertrophy, decreased cell shortening and prolonged relengthening duration, the effects of which were mitigated by ALDH2. Lectin staining displayed that diabetes promoted cardiac hypertrophy, the effect of which was alleviated by ALDH2. Western blot analysis revealed dampened autophagy protein markers including LC3B ratio and Atg7 along with upregulated p62 following experimental diabetes, the effect of which was reconciled by ALDH2. Phosphorylation level of AMPK was decreased and its downstream signaling molecule FOXO3a was upregulated in both diabetic cardiac tissue and in H9C2 cells with high glucose exposure. All these effect were partly abolished by ALDH2 overexpression and ALDH2 agonist Alda1. High glucose challenge dampened autophagy in H9C2 cells as evidenced by enhanced p62 levels and decreased levels of Atg7 and LC3B, the effect of which was alleviated by the ALDH2 activator Alda-1. High glucose-induced cell death and apoptosis were reversed by Alda-1. The autophagy inhibitor 3-MA and the AMPK inhibitor compound C mitigated Alda-1-offered beneficial effect whereas the autophagy inducer rapamycin mimicked or exacerbated high glucose-induced cell injury. Moreover, compound C nullified Alda-1-induced protection against STZ-induced changes in autophagy and function. Our results suggested that ALDH2 protects against diabetes-induced myocardial dysfunction possibly through an AMPK -dependent regulation of autophagy. This article is part of a Special Issue entitled: Autophagy and protein quality control in cardiometabolic diseases.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aldehyde Dehydrogenase/metabolism , Autophagy , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 1/physiopathology , Heart/physiopathology , Mitochondria/enzymology , Adenine/analogs & derivatives , Adenine/pharmacology , Aldehyde Dehydrogenase, Mitochondrial , Animals , Apoptosis/drug effects , Autophagy/drug effects , Autophagy-Related Protein-1 Homolog , Benzamides/pharmacology , Benzodioxoles/pharmacology , Calcium Signaling/drug effects , Cardiotonic Agents/metabolism , Cell Survival/drug effects , Chickens , Diabetes Mellitus, Type 1/pathology , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Glucose/pharmacology , Heart/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice, Transgenic , Mitochondria/drug effects , Models, Biological , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Rats , Streptozocin
6.
Biochim Biophys Acta ; 1852(2): 332-42, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24793415

ABSTRACT

Prolonged caloric restriction often results in alteration in heart geometry and function although the underlying mechanism remains poorly defined. Autophagy, a conserved pathway for bulk degradation of intracellular proteins and organelles, preserves energy and nutrient in the face of caloric insufficiency. This study was designed to examine the role of AMPK in prolonged caloric restriction-induced change in cardiac homeostasis and the underlying mechanism(s) involved with a focus on autophagy. Wild-type (WT) and AMPK kinase dead (KD) mice were caloric restricted (by 40%) for 30 weeks. Echocardiographic, cardiomyocyte contractile and intracellular Ca²âº properties, autophagy and autophagy regulatory proteins were evaluated. Caloric restriction compromised echocardiographic indices (decreased ventricular mass, left ventricular diameters, and cardiac output), cardiomyocyte contractile and intracellular Ca²âº properties associated with upregulated autophagy (Beclin-1, Atg5 and LC3BII-to-LC3BI ratio), increased autophagy adaptor protein p62, elevated phosphorylation of AMPK and TSC1/2, depressed phosphorylation of mTOR and ULK1. Although AMPK inhibition did not affect cardiac mechanical function, autophagy and autophagy signaling proteins, it significantly accentuated caloric restriction-induced changes in myocardial contractile function and intracellular Ca²âº handling. Interestingly, AMPK inhibition reversed caloric restriction-induced changes in autophagy and autophagy signaling. AMPK inhibition led to dampened levels of Beclin-1, Atg 5 and LC3B ratio along with suppressed phosphorylation of AMPK and TSC1/2 as well as elevated phosphorylation of mTOR and ULK1. Taken together, these data suggest an indispensible role for AMPK in the maintenance of cardiac homeostasis under prolonged caloric restriction-induced pathological changes possibly through autophagy regulation. This article is part of a Special Issue entitled: Autophagy and protein quality control in cardiometabolic diseases.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Autophagy , Caloric Restriction , Myocardial Contraction , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy/drug effects , Biological Transport/drug effects , Calcium/metabolism , Echocardiography , Glucose/metabolism , Glucose Tolerance Test , Male , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Myocardial Contraction/drug effects , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects
7.
Life Sci ; 94(1): 83-91, 2014 Jan 14.
Article in English | MEDLINE | ID: mdl-24269213

ABSTRACT

AIMS: Accumulation of advanced glycation endproduct (AGE) contributes to diabetic complication including diabetic cardiomyopathy although the precise underlying mechanism still remains elusive. Recent evidence depicted a pivotal role of protein kinase C (PKC) in diabetic complications. To this end, this study was designed to examine if PKCßII contributes to AGE-induced cardiomyocyte contractile and intracellular Ca(2+) aberrations. MAIN METHODS: Adult rat cardiomyocytes were incubated with methylglyoxal-AGE (MG-AGE) in the absence or presence of the PKCßII inhibitor LY333531 for 12h. Contractile and intracellular Ca(2+) properties were assessed using an IonOptix system including peak shortening (PS), maximal velocity of shortening/relengthening (±dL/dt), time-to-PS (TPS), time-to-90% relengthening (TR90), rise in intracellular Ca(2+) Fura-2 fluorescence intensity and intracellular Ca(2+) decay. Oxidative stress, O2(-) production and mitochondrial integrity were examined using TBARS, fluorescence imaging, aconitase activity and Western blotting. KEY FINDINGS: MG-AGE compromised contractile and intracellular Ca(2+) properties including reduced PS, ±dL/dt, prolonged TPS and TR90, decreased electrically stimulated rise in intracellular Ca(2+) and delayed intracellular Ca(2+) clearance, the effects of which were ablated by the PKCßII inhibitor LY333531. Inhibition of PKCßII rescued MG-AGE-induced oxidative stress, O2(-) generation, cell death, apoptosis and mitochondrial injury (reduced aconitase activity, UCP-2 and PGC-1α). In vitro studies revealed that PKCßII inhibition-induced beneficial effects were replicated by the NADPH oxidase inhibitor apocynin and were mitigated by the mitochondrial uncoupler FCCP. SIGNIFICANCE: These findings implicated the therapeutic potential of specific inhibition of PKCßII isoform in the management of AGE accumulation-induced myopathic anomalies.


Subject(s)
Glycation End Products, Advanced/metabolism , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Protein Kinase C beta/antagonists & inhibitors , Pyruvaldehyde/metabolism , Aconitate Hydratase/metabolism , Animals , Apoptosis , Blotting, Western , Calcium/metabolism , Indoles/pharmacology , Male , Maleimides/pharmacology , Mitochondria/pathology , Myocytes, Cardiac/pathology , Oxidative Stress , Rats , Rats, Sprague-Dawley
11.
BMC Med ; 10: 40, 2012 04 23.
Article in English | MEDLINE | ID: mdl-22524197

ABSTRACT

BACKGROUND: Mitochondrial aldehyde dehydrogenase (ALDH2) displays some promise in the protection against cardiovascular diseases although its role in diabetes has not been elucidated. METHODS: This study was designed to evaluate the impact of ALDH2 on streptozotocin-induced diabetic cardiomyopathy. Friendly virus B(FVB) and ALDH2 transgenic mice were treated with streptozotocin (intraperitoneal injection of 200 mg/kg) to induce diabetes. RESULTS: Echocardiographic evaluation revealed reduced fractional shortening, increased end-systolic and -diastolic diameter, and decreased wall thickness in streptozotocin-treated FVB mice. Streptozotocin led to a reduced respiratory exchange ratio; myocardial apoptosis and mitochondrial damage; cardiomyocyte contractile and intracellular Ca2+ defects, including depressed peak shortening and maximal velocity of shortening and relengthening; prolonged duration of shortening and relengthening; and dampened intracellular Ca2+ rise and clearance. Western blot analysis revealed disrupted phosphorylation of Akt, glycogen synthase kinase-3ß and Foxo3a (but not mammalian target of rapamycin), elevated PTEN phosphorylation and downregulated expression of mitochondrial proteins, peroxisome proliferator-activated receptor γ coactivator 1α and UCP-2. Intriguingly, ALDH2 attenuated or ablated streptozotocin-induced echocardiographic, mitochondrial, apoptotic and myocardial contractile and intracellular Ca2+ anomalies as well as changes in the phosphorylation of Akt, glycogen synthase kinase-3ß, Foxo3a and phosphatase and tensin homologue on chromosome ten, despite persistent hyperglycemia and a low respiratory exchange ratio. In vitro data revealed that the ALDH2 activator Alda-1 and glycogen synthase kinase-3ß inhibition protected against high glucose-induced mitochondrial and mechanical anomalies, the effect of which was cancelled by mitochondrial uncoupling. CONCLUSIONS: In summary, our data revealed that ALDH2 acted against diabetes-induced cardiac contractile and intracellular Ca2+ dysregulation, possibly through regulation of apoptosis, glycogen synthase kinase-3ß activation and mitochondrial function independent of the global metabolic profile.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/prevention & control , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/prevention & control , Glycogen Synthase Kinase 3/metabolism , Mitochondria/enzymology , Aldehyde Dehydrogenase, Mitochondrial , Animals , Apoptosis/drug effects , Calcium/metabolism , Diabetes Mellitus, Experimental/diagnostic imaging , Diabetes Mellitus, Experimental/physiopathology , Diabetic Cardiomyopathies/diagnostic imaging , Diabetic Cardiomyopathies/physiopathology , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Glucose/pharmacology , Glycogen Synthase Kinase 3 beta , Intracellular Space/drug effects , Intracellular Space/metabolism , Ion Channels/metabolism , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Oxygen Consumption/drug effects , PTEN Phosphohydrolase/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Proto-Oncogene Proteins c-akt/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sodium-Calcium Exchanger/metabolism , TOR Serine-Threonine Kinases/metabolism , Trans-Activators/metabolism , Transcription Factors , Ultrasonography , Uncoupling Protein 2
12.
J Cell Mol Med ; 13(8B): 1751-1764, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19602045

ABSTRACT

Diabetic cardiomyopathy is manifested by compromised systolic and diastolic function. This study was designed to examine the role of advanced glycation endproduct (AGE) and AGE receptor (RAGE) in diabetic cardiomyopathy. Heart function was assessed in isolated control and streptozotocin-induced diabetic hearts following in vivo RAGE gene knockdown using RNA interference. Cardiomyocyte mechanical properties were evaluated including peak shortening (PS), time-to-PS (TPS) and time-to-90% relengthening (TR(90)). RAGE was assayed by RT-PCR and immunoblot. Diabetes significantly enhanced cardiac MG, AGE and RAGE levels accompanied with colocalization of AGE and RAGE in cardiomyocytes. Diabetes-elicited increase in RAGE was inhibited by in vivo siRNA interference. The AGE formation inhibitor benfotiamine significantly attenuated diabetes-induced elevation in MG, AGE, RAGE and collagen cross-linking without affecting hypertriglyceridaemia and hypercholesterolaemia in diabetes. Diabetes markedly decreased LV contractility, as evidenced by reduced +/-dP/dt and LV developed pressure (LVDP), which were protected by RAGE gene knockdown. In addition, MG-derived AGE (MG-AGE) up-regulated cardiac RAGE mRNA and triggered cardiomyocyte contractile dysfunction reminiscent of diabetic cardiomyopathy. The MG-AGE-elicited prolongation of TPS and TR(90) was ablated by an anti-RAGE antibody in cardiomyocytes. Interestingly, MG-AGE-induced cardiomyocyte dysfunction was associated with mitochondrial membrane potential (MMP) depolarization and reduced GSK-3beta inactivation in control cardiomyocytes, similar to those from in vivo diabetes. Treatment with siRNA-RAGE ablated diabetes-induced MMP depolarization and GSK-3beta inactivation. Collectively, our result implicated a role of AGE-RAGE in the pathogenesis of diabetic cardiomyopathy.


Subject(s)
Cardiomyopathies/metabolism , Diabetes Mellitus, Experimental/metabolism , Glycation End Products, Advanced/metabolism , Receptors, Immunologic/metabolism , Up-Regulation , Animals , Diabetes Mellitus, Experimental/complications , Enzyme-Linked Immunosorbent Assay , Gene Knockdown Techniques , Male , Mice , RNA Interference , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Streptozocin
13.
Toxicol Lett ; 187(3): 149-56, 2009 Jun 22.
Article in English | MEDLINE | ID: mdl-19429258

ABSTRACT

Chronic intake of alcohol results in multiple organ damage including brain. This study was designed to examine the impact of facilitated acetaldehyde breakdown via transgenic overexpression of mitochondrial aldehyde dehydrogenase-2 (ALDH2) on alcohol-induced cerebral cortical injury. ALDH2 transgenic mice were produced using the chicken beta-actin promoter. Wild-type FVB and ALDH2 mice were placed on a 4% alcohol or control diet for 12 weeks. Protein damage and apoptosis were evaluated with carbonyl formation, caspase and TUNEL assays. Western blot was performed to examine expression (or its activation) of ALDH2, the pro- and anti-apoptotic proteins caspase-8, Bax, Bcl-2, Omi/HtrA2, apoptosis repressor with caspase recruitment domain (ARC), FLICE-like inhibitory protein (FLIP), X-linked inhibitor of apoptosis protein (XIAP), Akt, glycogen synthase kinase-3beta (GSK-3beta), p38, c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK). Chronic alcohol intake led to elevated apoptosis in the absence of overt protein damage, the effect of which was ablated by the overexpression of ALDH2 transgene. Consistently, ALDH2 transgene significantly attenuated alcohol-induced upregulation of Bax, Omi/HtrA2 and XIAP as well as downregulation of Bcl-2 and ARC without affecting alcohol-induced increase of FLIP in cerebral cortex. Phosphorylation of Akt and GSK-3beta was dampened while total/phosphorylated JNK and p38 phosphorylation were elevated following chronic alcohol intake, the effects of which were abrogated by ALDH2 transgene. Expression of total Akt, GSK-3beta, p38 and ERK (total or phosphorylated) was not affected by either chronic alcohol intake or ALDH2 transgene. Our results suggested that transgenic overexpression of ALDH2 rescues chronic alcoholism-elicited cerebral injury possibly via a mechanism associated with Akt, GSK-3beta, p38 and JNK signaling.


Subject(s)
Alcohol Drinking/metabolism , Aldehyde Dehydrogenase/metabolism , Apoptosis/drug effects , Cerebral Cortex/metabolism , Acetaldehyde/metabolism , Alcohol Drinking/pathology , Aldehyde Dehydrogenase/biosynthesis , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase, Mitochondrial , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins/biosynthesis , Blotting, Western , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Ethanol/administration & dosage , Ethanol/toxicity , Female , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , In Situ Nick-End Labeling , Male , Mice , Mice, Transgenic , Protein Carbonylation/physiology , Proto-Oncogene Proteins c-akt/metabolism
14.
Hypertension ; 52(6): 1161-7, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18981324

ABSTRACT

Reductions in uterine perfusion pressure (RUPP) in pregnant rats is associated with increased tumor necrosis factor-alpha (TNF-alpha). This study was designed to determine the role of endogenous TNF-alpha in mediating changes in arterial pressure and endothelin-1 (ET-1) in RUPP rats. To achieve this goal we examined the effect of RUPP in the presence and absence of a TNF-alpha-soluble receptor, etanerecept (0.4 mg/kg). Mean arterial pressure increased from 102+/-1 mm Hg in normal pregnant (NP) rats to 134+/-3 mm Hg (P<0.05) in RUPP rats. Serum TNF-alpha increased to 40+/-7.6 pg/mL in RUPP rats (n=24) versus 14.8+/-3.3 pg/mL (n=16; P<0.05) in NP rats. Administration of etanerecept decreased TNF-alpha in RUPP rats (n=20) to 17.2+/-3 pg/mL and mean arterial pressure to 118+/-2 mm Hg (P<0.05). Tissue ET-1 decreased in etanerecept-treated RUPP rats compared with control RUPP rats. The direct effect of TNF-alpha blockade on endothelial activation in response to placental ischemia was examined in human umbilical vein endothelial cells. ET-1 secreted from human umbilical vein endothelial cells treated with RUPP serum was 59.2+16 pg/mg and decreased when etanerecept was added to the medium with RUPP serum (7.60+/-0.77 pg/mg), as well as in response to serum from etanerecept-treated RUPP rats (7.30+/-0.55 pg/mg; P<0.001). ET-1 secreted from human umbilical vein endothelial cells was 15.6+/-2 pg/mg when treated with NP serum. These data support the hypothesis that endogenous TNF-alpha is an important stimulus for ET-1 in response to placental ischemia and is important in mediating endothelial cell activation and hypertension during pregnancy.


Subject(s)
Blood Pressure/physiology , Hypertension, Pregnancy-Induced/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/blood , Uterus/blood supply , Animals , Birth Weight , Blood Pressure/drug effects , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelin-1/genetics , Endothelin-1/metabolism , Etanercept , Female , Humans , Hypertension, Pregnancy-Induced/physiopathology , Immunoglobulin G/pharmacology , Immunologic Factors/pharmacology , Ischemia/metabolism , Ischemia/physiopathology , Kidney/physiology , Organ Size , Placenta/anatomy & histology , Placenta/physiology , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor-alpha/genetics , Umbilical Veins/cytology , Uterus/physiology
15.
Hypertension ; 50(6): 1142-7, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17923588

ABSTRACT

The balance between proangiogenic and antiangiogenic factors, such as vascular endothelial growth factor, placental growth factor, and soluble fms-like tyrosine kinase-1 (sFlt-1), is altered in preeclampsia, and this dysregulation of angiogenic factors may be important in the pathogenesis of preeclampsia. Although sFlt-1 is elevated in preeclampsia, the mechanisms responsible for increasing this antiangiogenic factor remain unclear. We hypothesized that the hypertension produced by reduced uterine perfusion pressure (RUPP) is associated with increased sFlt-1 expression and decreased plasma vascular endothelial growth factor and placental growth factor concentrations in the pregnant rat. Arterial pressure was increased (130+/-3 versus 100+/-2 mm Hg; P<0.01) in the RUPP rats compared with the normal pregnant control rats. Plasma sFlt-1 concentration (660+/-270 versus 82+/-26 pg/mL; P<0.05) was increased, whereas plasma free placental growth factor (0.28+/-0.05 versus 1.7+/-0.5 pg/mL; P<0.01) and vascular endothelial growth factor (594+/-34 versus 830+/-33 pg/mL; P<0.01) concentrations were decreased in the RUPP rats compared with normal pregnant rats. Plasma sFlt-1:placental growth factor (37.2+/-7.8 versus 8.9+/-1.6; P<0.02) and sFlt-1:vascular endothelial growth factor (0.86+/-0.22 versus 0.28+/-0.06; P<0.05) ratios were increased in the RUPP rats compared with normal pregnant rats. Immunoreactive placental sFlt-1 was increased (1.1+/-0.1 versus 0.3+/-0.1; P<0.01) in RUPP rats contrasted with the normal pregnant rats. These findings support our hypothesis that RUPP increases the expression of sFlt-1 and alters the balance of angiogenic factors in the maternal circulation. These data also indicate that the RUPP model of pregnancy-induced hypertension may provide an invaluable model for mechanistic studies into the role of sFlt-1 in the pathogenesis preeclampsia.


Subject(s)
Pre-Eclampsia/etiology , Uterus/blood supply , Vascular Endothelial Growth Factor Receptor-1/physiology , Amniotic Fluid/chemistry , Animals , Disease Models, Animal , Female , Fetal Development , Placenta Growth Factor , Pregnancy , Pregnancy Proteins/blood , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor Receptor-1/analysis
16.
Pediatr Res ; 61(1): 42-7, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17211139

ABSTRACT

Inadequate nutrition compromises fetal development and poses long-term health risks for the offspring, even without decreased birth weight. The present study sought to 1) establish the ontogeny of fetal renal glomerulus number (GN) in sheep and 2) evaluate the effects of 50% global nutrient restriction (NR) during early to midgestation on GN and the renin-angiotensin system in the fetal kidney. GN increased from 78 dG (68,560 +/- 3802) to 135 dG (586,118 +/- 25,792). NR increased combined kidney weight (29 +/- 0.6 g versus 23 +/- 1.1 g), whereas decreased GN relative to right kidney weight approached significance in males (26,000 +/- 5300 versus 39,000 +/- 2800 GN/g) compared with control (C) males and females. NR decreased immunoreactive angiotensin II (Ang II) type 1 receptor (AT1) in the NR kidneys at 78 dG and increased renin at 135 dG. Immunoreactive renin decreased from 78 to 135 dG. Female fetuses had more immunoreactive Ang II type 2 receptor (AT2) than male fetuses at 78 dG and males had more AT1 at 135 dG. The present study demonstrates gender-specific differences in fetal growth and development and in fetal kidney development in pregnancies affected by NR.


Subject(s)
Eating/physiology , Food , Kidney Glomerulus/embryology , Animals , Female , Fetus , Male , Pregnancy , Sex Factors , Sheep
17.
Mech Ageing Dev ; 128(3): 276-85, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17250874

ABSTRACT

Catalase, an enzyme which detoxifies H2O2, may interfere with cardiac aging. To test this hypothesis, contractile and intracellular Ca2+ properties were evaluated in cardiomyocytes from young (3-4 months) and old (26-28 months) FVB and transgenic mice with cardiac overexpression of catalase. Contractile indices analyzed included peak shortening (PS), time-to-90% PS (TPS90), time-to-90% relengthening (TR90), half-width duration (HWD), maximal velocity of shortening/relengthening (+/-dL/dt) and intracellular Ca2+ levels or decay rate. Levels of advanced glycation endproduct (AGE), Na+/Ca2+ exchanger (NCX), sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA2a), phospholamban (PLB), myosin heavy chain (MHC), membrane Ca2+ and K+ channels were measured by western blot. Catalase transgene prolonged survival while did not alter myocyte function by itself. Aging depressed+/-dL/dt, prolonged HWD, TR90 and intracellular Ca2+ decay without affecting other indices in FVB myocytes. Aged FVB myocytes exhibited a stepper decline in PS in response to elevated stimulus or a dampened rise in PS in response to elevated extracellular Ca2+ levels. Interestingly, aging-induced defects were nullified or significantly attenuated by catalase. AGE level was elevated by 5-fold in aged FVB compared with young FVB mice, which was reduced by catalase. Expression of SERCA2a, NCX and Kv1.2 K+ channel was significantly reduced although levels of PLB, L-type Ca2+ channel dihydropyridine receptor and beta-MHC isozyme remained unchanged in aged FVB hearts. Catalase restored NCX and Kv1.2 K+ channel but not SERCA2a level in aged mice. In summary, our data suggested that catalase protects cardiomyocytes from aging-induced contractile defect possibly via improved intracellular Ca2+ handling.


Subject(s)
Aging/physiology , Antioxidants/metabolism , Cardiomyopathies/enzymology , Cardiomyopathies/physiopathology , Catalase/biosynthesis , Myocardial Contraction/physiology , Myocardium/enzymology , Myocytes, Cardiac/physiology , Animals , Blotting, Western , Calcium Channels, L-Type/metabolism , Calcium Signaling/physiology , Calcium-Binding Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Glycation End Products, Advanced/metabolism , Isoenzymes/metabolism , Mice , Mice, Transgenic , Myosin Heavy Chains/metabolism , Oxidative Stress/physiology , Peroxisomes/enzymology , Potassium Channels/metabolism , Protein Processing, Post-Translational/physiology , Sarcoplasmic Reticulum/enzymology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sodium-Calcium Exchanger/metabolism , Survival
18.
Life Sci ; 80(11): 1051-6, 2007 Feb 20.
Article in English | MEDLINE | ID: mdl-17174344

ABSTRACT

Accumulation of advanced glycation endproduct (AGE) has been implicated in the pathogenesis of diabetic complications. However, the precise role and mechanism behind AGE-associated diabetic heart injury are not fully clear. This study was designed to evaluate the effect of AGE on accumulation of reactive oxygen species (ROS), apoptosis, mitogen-activated protein kinase (MAPK) activation and nuclear O-GlcNAcylation in fetal human cardiac myocytes. Myocytes were maintained for 24-72 h in a defined culture medium containing high glucose, the AGE carbon precursor methylglyoxal (MG), and MG-AGE derived from MG and bovine serum albumin (BSA). Generation of ROS was detected by 5-(6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate. Apoptosis was evaluated by caspase-3 activity and quantitative DNA fragmentation. Both high glucose (25.5 mM) and MG (200 microM) significantly enhanced ROS and AGE formation with greater effects elicited by MG. Both high glucose and MG-AGE significantly facilitated apoptosis with a more predominant effect from MG-AGE. In addition, phosphorylation of MAPK cascade [extracellular signal-regulated kinase-1/2 (ERK1/2) and p38] and nuclear O-GlcNAcylation were enhanced in MG-AGE-treated myocytes, similar to those elicited by high glucose. MG-AGE-induced phosphorylation of ERK1/2 and p38 was nullified by neutralizing AGE with specific anti-AGE antibody but not nonspecific antiserum. Collectively, these results indicated that AGE or its precursor MG may trigger ROS generation, apoptosis, MAPK activation and nuclear O-GlcNAcylation in human cardiac myocytes, in a manner reminiscent of high extracellular glucose.


Subject(s)
Apoptosis/drug effects , Glycation End Products, Advanced/pharmacology , Mitogen-Activated Protein Kinase Kinases/biosynthesis , Myocytes, Cardiac/pathology , N-Acetylglucosaminyltransferases/metabolism , Reactive Oxygen Species/metabolism , Acylation , Animals , Caspase 3/metabolism , Cattle , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , DNA Fragmentation , Fetus/cytology , Glucose/pharmacology , Glycosylation , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Pyruvaldehyde/pharmacology , Serum Albumin, Bovine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...