Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 4616, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36944665

ABSTRACT

Oxytocin is a hormone with functions in: reproduction, maternal bonding, milk ejection, and feeding/social behavior, and is reported to be present in a variety of tissues. Our goal is to characterize oxytocin and leucyl and cystinyl aminopeptidase (LNPEP/oxytocinase), a key regulator of oxytocin in mares. We measured serum and tissue LNPEP by ELISA from ovulation (D0) until D21-22 in non-pregnant (n = 5) and pregnant mares (n = 6); and in periparturient and postpartum mares (n = 18). Placenta (n = 7) and homogenized tissue of diestrus mares (n = 6) were evaluated using protein determinations and LNPEP ELISAs. Identification of LNPEP and OXT protein in tissues was also performed via western blot, immunohistochemistry and liquid chromatography-mass spectrometry (LC-MS/MS). Furthermore, in situ hybridization was performed for LNPEP and OXT on endometrium, myometrium, pituitary and corpus luteum (CL). Serum LNPEP concentration were similar. Placental LNPEP U/mg protein was highest in the body and pregnant horn. The highest to lowest LNPEP U/mg protein by tissue were: myometrium > follicle wall > endometrium > kidney > CL > liver. Oxytocin was identified in the equine pituitary, CL and placenta and is likely to act in autocrine or paracrine manner, while LNPEP may act systemically and locally to regulate the availability of OXT.


Subject(s)
Cystinyl Aminopeptidase , Oxytocin , Horses , Animals , Female , Pregnancy , Oxytocin/metabolism , Cystinyl Aminopeptidase/metabolism , Placenta/metabolism , Chromatography, Liquid , Tandem Mass Spectrometry
2.
Cureus ; 13(10): e18661, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34786247

ABSTRACT

Background Congestive heart failure (CHF) readmissions are associated with substantial financial and medical implications. We performed a descriptive study to determine demographic, clinical, and behavioral factors associated with 30-day readmission. Materials and methods Patients hospitalized with CHF at William Beaumont Hospital in Royal Oak, MI, from March 2019-May 2019 were studied. Response to heart failure knowledge and self-care questionnaires along with the patients' demographic and clinical factors were collected. Thirty-day readmission to any of the eight hospitals in the Beaumont Health System was documented. Results One-hundred ninety-six (196) patients were included. The all-cause 30-day readmission rate was 23%. A numerical higher rate of readmissions was observed among males (23.7% vs 22.2%), current smokers (27.3% vs 22.9%), and patients with peripheral vascular disease (PVD; 28.9% vs 21.2%), diabetes mellitus (DM; 26.4% vs 18.9%), hypertension (HTN; 26.4% vs 10%), coronary artery disease (CAD; 24.6% vs 19%), and prior history of cerebrovascular accident (CVA; 28.9% vs 21.2%) (p>0.05). Reduced left ventricular ejection fraction (LVEF) was associated with higher readmissions (24.4% vs 20.5%, p=0.801). Patients with the highest reported questionnaire scores corresponding to better heart failure knowledge and self-care behaviors at home were readmitted at a similar rate compared to those scoring in the lowest interval (25%, p=0.681). Conclusion Though statistically insignificant due to the limitations of sample size, a higher percentage of readmissions was observed in male patients, current smokers, reduced LVEF, and higher comorbidity burden. Better reported patient self-care behavior, medication compliance, and heart failure knowledge did not correlate with reduced readmission rates. While the impact of medical comorbidities on 30-day readmissions is better established, the role of socioeconomic factors remains unclear and might suggest a focus for future work.

4.
Clin Exp Pharmacol Physiol ; 44(10): 1008-1016, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28636787

ABSTRACT

Using our in vitro and in vivo models of oxidative stress, the current study was designed to determine the neuroprotective potential of naringenin, alone or in combination with lipoic acid. In our mixed neuronal culture exposed to hypoxia and subsequent reoxygenation, naringenin was shown to provide significant neuroprotection against cell death at a concentration of 2.5 µmol/L. Lipoic acid (LA) did not produce neuroprotection at any concentration tested (0.25-100 µmol/L). In contrast, when naringenin was covalently combined with LA, producing a novel compound named "VANL-100", significant neuroprotection was observed at a concentration as low as 2×10-2  µmol/L (100-fold more potent). An ELISA for antioxidant capacity demonstrated that naringenin and VANL-100 likely resulted in neuroprotection by increasing the free radical scavenging capacity of the neuronal cells. Pretreatment of rats with the above compounds prior to middle cerebral artery occlusion (MCAO) followed by reperfusion, showed similar results. Naringenin significantly reduced infarct volume at a dose of 10 mg/kg while VANL-100 produced significant neuroprotection at a dose as low as 1×10-4  mg/kg (10 000-fold more potent). This VANL-100-induced neuroprotection persisted even when administered 1 and 3 hours into the reperfusion time course. Taken together, these results suggest that our novel compound, VANL-100 is neuroprotective, likely via a mechanism that involves increasing the antioxidant capacity of neuronal cells. Our results also show that VANL-100 is 100-10 000-fold more potent than the parent compounds, which adds to the growing evidence in support of combination therapy targeting oxidative stress in neurodegenerative diseases.


Subject(s)
Flavanones/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Thioctic Acid/pharmacology , Animals , Antioxidants/metabolism , Disease Models, Animal , Female , Flavanones/administration & dosage , Flavanones/therapeutic use , Glucose/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Oxygen/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Thioctic Acid/administration & dosage , Thioctic Acid/therapeutic use
5.
Clin Exp Pharmacol Physiol ; 44(10): 993-1000, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28504843

ABSTRACT

In this study, we tested a novel synthetic pyrazole-containing compound, 5-amino-1-phenyl-1H-pyrazole-4-carbonitrile (APPC), as an antioxidant in both in vitro and in vivo models of oxidative stress. In addition, the utility of covalently combining APPC with another well-established antioxidant, lipoic acid (LA), was also tested in both models. The in vitro results demonstrated that pretreatment with APPC in a mixed neuronal-glial culture exposed to oxygen-glucose deprivation (OGD) followed by reoxygenation-refeeding, resulted in significant neuroprotection at concentrations between 2.5 to 25 µmol/L. In contrast, LA was not neuroprotective following OGD alone or following reoxygenation-refeeding. However, the synthetic covalent combination of APPC with LA, named "UPEI-800", resulted in significant neuroprotection at concentrations between 0.027 and 2.7 µmol/L (100-fold more potent than APPC alone), an effect shown to be correlated with increased cellular antioxidant capacity. Further, in an in vivo model of ischaemia-reperfusion injury following transient occlusion of the middle cerebral artery (tMCAO), both APPC (0.1 and 1.0 mg/kg) and UPEI-800 (1×10-3  mg/kg) provided significant neuroprotection. Consistent with the in vitro findings, the in vivo results following tMCAO also demonstrated a 100-fold increase in the potency of the covalently linked compound UPEI-800 compared to APPC alone.


Subject(s)
Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Pyrazoles/pharmacology , Animals , Antioxidants/metabolism , Cell Death/drug effects , Chemistry Techniques, Synthetic , Glucose/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Oxygen/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Rats , Reperfusion Injury/pathology
6.
Food Chem Toxicol ; 100: 175-182, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28017701

ABSTRACT

Previously, our laboratory provided evidence that lipoic acid (LA) covalently bonded to various antioxidants, resulted in enhanced neuroprotection compared to LA on its own. The naturally occurring compound scopoletin, a coumarin derivative, has been shown in various in vitro studies to have both antioxidant and anti-inflammatory mechanism of actions. The present investigation was designed to determine if scopoletin on its own, or a co-drug consisting of LA and scopoletin covalently bonded together, named UPEI-400, would be capable of demonstrating a similar neuroprotective efficacy. Using a rat stroke model, male rats were anesthetized (Inactin®; 100 mg/kg, iv), the middle cerebral artery was permanently occluded for 6 h (pMCAO), or in separate animals, occluded for 30 min followed by 5.5 h of reperfusion (ischemia/reperfusion; I/R). Pre-administration of either scopoletin or UPEI-400 significantly decreased infarct volume in the I/R model (p < 0.05), but not in the pMCAO model of stroke. UPEI-400 was ∼1000 times more potent compared to scopoletin alone. Since UPEI-400 was only effective in a model of I/R, it is possible that it may act to enhance neuronal antioxidant capacity and/or upregulate anti-inflammatory pathways to prevent the neuronal cell death.


Subject(s)
Brain Ischemia/drug therapy , Disease Models, Animal , Neuroprotective Agents/pharmacology , Reperfusion Injury/prevention & control , Scopoletin/analogs & derivatives , Scopoletin/pharmacology , Stroke/prevention & control , Thioctic Acid/analogs & derivatives , Thioctic Acid/pharmacology , Animals , Male , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley , Scopoletin/administration & dosage , Thioctic Acid/administration & dosage
7.
PLoS One ; 9(1): e87865, 2014.
Article in English | MEDLINE | ID: mdl-24498217

ABSTRACT

The present study demonstrates the benefits of combinatorial antioxidant therapy in the treatment of ischemic stroke. Male Sprague-Dawley rats were anaesthetised and the middle cerebral artery (MCA) was occluded for 30 minutes followed by 5.5 hours of reperfusion. Pretreatment with resveratrol 30 minutes prior to MCA occlusion resulted in a significant, dose-dependent decrease in infarct volume (p<0.05) compared to vehicle-treated animals. Neuroprotection was also observed when resveratrol (2 × 10(-3) mg/kg; iv) was administered within 60 minutes following the return of blood flow (reperfusion). Pretreatment with non-neuroprotective doses of resveratrol (2 × 10(-6) mg/kg) and lipoic acid (LA; 0.005 mg/kg) in combination produced significant neuroprotection as well. This neuroprotection was also observed when resveratrol and LA were administered 15 minutes following the onset of MCA occlusion. Subsequently, we synthetically combined resveratrol and LA in both a 1 ∶ 3 (UPEI-200) and 1 ∶ 1 (UPEI-201) ratio, and screened these new chemical entities in both permanent and transient ischemia models. UPEI-200 was ineffective, while UPEI-201 demonstrated significant, dose-dependent neuroprotection. These results demonstrate that combining subthreshold doses of resveratrol and LA prior to ischemia-reperfusion can provide significant neuroprotection likely resulting from concurrent effects on multiple pathways. The additional protection observed in the novel compound UPEI 201 may present opportunities for addressing ischemia-induced damage in patients presenting with transient ischemic episodes.


Subject(s)
Antioxidants/pharmacology , Brain Ischemia/drug therapy , Reperfusion Injury/drug therapy , Stilbenes/pharmacology , Stroke/drug therapy , Thioctic Acid/pharmacology , Animals , Brain Ischemia/physiopathology , Disease Models, Animal , Humans , Male , Rats , Rats, Sprague-Dawley , Reperfusion Injury/physiopathology , Resveratrol , Stroke/physiopathology
8.
Neurosci Lett ; 561: 151-5, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24394910

ABSTRACT

Edaravone, an electron spin trapper with radical scavenging activity, has been shown to be effective in reducing infarct volume in humans following ischemic stroke. However, concerns of edaravone-induced renal toxicity have limited its clinical adoption. Previous work has demonstrated that edaravone produced significant neuroprotection when injected prior to a period of ischemia and/or reperfusion. The current investigation was designed to determine if a newly synthesized co-drug consisting of lipoic acid and edaravone, named UPEI-300, could produce neuroprotection in in vitro and/or an in vivo rodent model of stroke. UPEI-300 produced dose-dependent neuroprotection in vitro and was subsequently tested in vivo. Male rats were anaesthetized and the middle cerebral artery was occluded for 30 min followed by 5.5 h of reperfusion (ischemia/reperfusion; I/R). Pre-administration of UPEI-300 dose-dependently decreased infarct volume. Significant neuroprotection was also observed when UPEI-300 (1.0 mg/kg) was injected during the 30 min period of ischemia as well as up to 60 min following the start of reperfusion. These results indicate that a co-drug consisting of edaravone and lipoic acid is a potent neuroprotectant, and clinically, the use of such a novel co-drug following an ischemic stroke might maintain neuroprotection while potentially decreasing edaravone associated renal toxicity.


Subject(s)
Antipyrine/analogs & derivatives , Brain Ischemia/prevention & control , Neuroglia/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Reperfusion Injury/prevention & control , Stroke/prevention & control , Thioctic Acid/analogs & derivatives , Animals , Antipyrine/pharmacology , Antipyrine/therapeutic use , Brain Ischemia/pathology , Cell Hypoxia , Cells, Cultured , Drug Combinations , Male , Neocortex/cytology , Neuroglia/pathology , Neurons/pathology , Neuroprotective Agents/therapeutic use , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Stroke/pathology , Thioctic Acid/pharmacology , Thioctic Acid/therapeutic use
9.
Neurosci Lett ; 548: 217-21, 2013 Aug 26.
Article in English | MEDLINE | ID: mdl-23748073

ABSTRACT

Resveratrol, a dietary polyphenol with antioxidant and anti-inflammatory activity, has been shown to provide neuroprotection in models of ischemia. However, the mechanism of action of resveratrol-induced neuroprotection remains unclear. Previous work in our laboratory has provided evidence that acute, systemic administration of resveratrol is neuroprotective in a permanent model of cerebral ischemia, an effect that was blocked when animals received the non-selective estrogen receptor antagonist, ICI, 182,780. The present study was designed to investigate whether the source of neuroprotection afforded by resveratrol action within the cerebral cortex itself is mediated preferentially via selective activation of either α or ß estrogen receptor subtype. Intracortical injection of resveratrol (0.1 and 1.0 µM) 10 min prior to 30 min of ischemia followed by 5.5h of reperfusion significantly reduced infarct volume in the prefrontal cortex. This neuroprotective effect was significantly attenuated when resveratrol injection (1.0 µM) was preceded by injection of a selective estrogen receptor α antagonist, 1,3-bis(4-hydroxyphenyl)-4-methyl-5-[4-(2-piperidinylethoxy)phenol]-1N-pyrozole dihydrochloride (MPP) or a selective estrogen receptor beta (ERß) antagonist, 4-[2-phenyo-5,7-bis(trifluoromrthyl)pyrazolo(1,5-a)pyrimidin-3-yl]phenol (PHTPP). These results provide evidence for rapidly induced neuroprotection mediated by resveratrol activation of either estrogen receptor subtype within the ischemic cortex of rats.


Subject(s)
Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Stilbenes/administration & dosage , Animals , Brain Ischemia/diagnosis , Cerebral Cortex/blood supply , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Male , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley , Reperfusion Injury/diagnosis , Resveratrol , Treatment Outcome
10.
Dev Dyn ; 242(6): 790-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23559562

ABSTRACT

BACKGROUND: The DNA-binding transcription factor Wilms' Tumor Suppressor-1 (WT1) plays an essential role in nephron progenitor differentiation during renal development. We previously used Wt1 chromatin-immunoprecipitation coupled to microarray (ChIP-chip) to identify novel Wt1 target genes that may regulate nephrogenesis in vivo. We discovered that all three members of the SoxC subfamily, namely, Sox4, Sox11, and Sox12, are bound by Wt1 in mouse embryonic kidneys in vivo. SoxC genes play master roles in determining neuronal and mesenchymal progenitor cell fate in a multitude of developmental processes, but their function in the developing kidney is largely unknown. RESULTS: Here we show that all three SoxC genes are expressed in the nephrogenic lineages during renal development. Conditional ablation of Sox4 in nephron progenitors and their cellular descendants (Sox4(nephron-) mice) results in a significant reduction in nephron endowment. By postnatal day (P)7, Sox4(nephron-) renal corpuscles exhibit reduced numbers of Wt1+ podocytes together with loss of expression of the slit diaphragm protein nephrin. Sox4(nephron-) mice develop early-onset proteinacious glomerular injury within 2 weeks of birth progressing to end-stage renal failure within 5-9 months. CONCLUSIONS: Collectively, our results demonstrate an essential requirement of Sox4 for normal renal development in vivo.


Subject(s)
Gene Expression Regulation, Developmental , Kidney/embryology , SOXC Transcription Factors/metabolism , Alleles , Animals , Cell Lineage , Chromatin Immunoprecipitation , In Situ Hybridization , Kidney Glomerulus/metabolism , Mice , Microscopy, Electron, Transmission , Nephrons/metabolism , Oligonucleotide Array Sequence Analysis , Renal Insufficiency/genetics , Stem Cells/cytology , Time Factors , WT1 Proteins/metabolism
11.
J Neurosci Res ; 91(2): 262-72, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23151946

ABSTRACT

After ischemic stroke, early thrombolytic therapy to reestablish tissue perfusion improves outcome but triggers a cascade of deleterious cellular and molecular events. Using a collaborative approach, our groups examined the effects of guanosine (Guo) in response to ischemic reperfusion injury in vitro and in vivo. In a transient middle cerebral artery occlusion (MCAO) in rats, Guo significantly reduced infarct volume in a dose-dependent manner when given systemically either immediately before or 30 min, but not 60 min, after the onset of the 5.5-hr reperfusion period. In a separate experiment, Guo significantly reduced infarct volume after 24 hr of reperfusion when administered 5 min before reperfusion. Western blot analysis did not reveal any significant changes either in endoplasmic reticulum (ER) stress proteins (GRP 78 and 94) or HSP 70 or in levels of m-calpain. In vitro oxygen and glucose deprivation (OGD) significantly increased production of both reactive oxygen species (ROS) and interleukin-8 (IL-8) in the primary astrocytes. Guo did not alter ROS or IL-8 production when given to the astrocytes before OGD. However, Guo when added to the cells prior to or 30 min after reperfusion significantly reduced IL-8 release but not ROS formation. Our study revealed a dose- and time-dependent protective effect of Guo on reperfusion injury in vitro and vivo. The mechanisms by which Guo exerts its effect are independent of unfolded proteins in ER or the level of intracellular calcium or ROS formation. However, the effect may be induced, at least partially, by inhibiting IL-8, a marker of reperfusion-triggered proinflammatory events.


Subject(s)
Brain Infarction/prevention & control , Guanosine/administration & dosage , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/administration & dosage , Reperfusion Injury/prevention & control , Analysis of Variance , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/metabolism , Brain Infarction/etiology , Cells, Cultured , Gene Expression Regulation/drug effects , Glucose/deficiency , Heat-Shock Proteins/metabolism , Hypoxia , Interleukin-8/metabolism , Male , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Reperfusion/adverse effects , Reperfusion Injury/complications , Time Factors
12.
Am J Physiol Regul Integr Comp Physiol ; 302(7): R886-95, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22277933

ABSTRACT

Previous work in our laboratory has provided evidence that preadministration of apocynin and lipoic acid at subthreshold levels for neuroprotection enhanced the neuroprotective capacity when injected in combination. Therefore, the present investigation was designed to determine whether a co-drug consisting of lipoic acid and apocynin functional groups bound by a covalent bond, named UPEI-100, is capable of similar efficacy using a rodent model of stroke. Male rats were anesthetized with Inactin (100 mg/kg iv), and the middle cerebral artery was occluded for 6 h or allowed to reperfuse for 5.5 h following a 30-min occlusion (ischemia/reperfusion, I/R). Preadministration of UPEI-100 dose-dependently decreased infarct volume in the I/R model (P < 0.05), but not in the middle cerebral artery occlusion model of stroke. Using the optimal dose, we then injected UPEI-100 during the stroke or at several time points during reperfusion, and significant neuroprotection was observed when UPEI-100 was administered up to 90 min following the start of reperfusion (P < 0.05). A time course for this neuroprotective effect showed that UPEI-100 resulted in a decrease in infarct volume following 2 h of reperfusion compared with vehicle. The time course of this neuroprotective effect was also used to study several mediators along the antioxidant pathway and showed that UPEI-100 increased the level of mitochondrial superoxide dismutase and oxidized glutathione and decreased a marker of lipid peroxidation due to oxidative stress (HNE-His adduct formation). Taken together, the data suggest that UPEI-100 may utilize similar pathways to those observed for the two parent compounds; however, it may also act through a different mechanism of action.


Subject(s)
Acetophenones/therapeutic use , Neuroprotective Agents/therapeutic use , Reperfusion Injury/drug therapy , Stroke/drug therapy , Thioctic Acid/analogs & derivatives , Thioctic Acid/therapeutic use , Acetophenones/chemical synthesis , Acetophenones/chemistry , Animals , Biomarkers/metabolism , Disease Models, Animal , Glutathione Disulfide/biosynthesis , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Lipid Peroxidation/drug effects , Male , Mitochondria/drug effects , Mitochondria/enzymology , Neuroprotective Agents/chemical synthesis , Oxidative Stress , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Stroke/metabolism , Stroke/prevention & control , Superoxide Dismutase/biosynthesis , Thioctic Acid/chemical synthesis , Thioctic Acid/chemistry
13.
Food Chem Toxicol ; 49(12): 3063-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21946070

ABSTRACT

The present study was designed to determine a dose-response relationship between apocynin and infarct volume as well as to provide a possible molecular mechanism mediating this effect. We tested the hypothesis that apocynin protects against cell death following stroke and reperfusion injury. Apocynin was administered 30 min prior to, or immediately following removal of sutures used to occlude the middle cerebral artery (MCA) in male Sprague-Dawley rats. Following removal of the sutures, the MCA was allowed to undergo 5.5h of reperfusion. Pretreatment with apocynin 30 min prior to occlusion resulted in a dose-dependent reduction in infarct volume by ∼50 %. Analysis of tissue from the ischemic cortex of apocynin-treated rats showed an increase in the level of glutathione (GSH), protein adducts (HNE-His), hydrogen peroxide (H(2)O(2)) and DNA fragmentation (apoptotic cell death) was also observed. This suggests that apocynin may increase antioxidant defense systems (GSH) to limit the degree of ischemia-induced cellular stress. In addition, this moderate cell stress results in more apoptotic vs necrotic cell death, and thus may limit the spreading depression and total cell death that occurs following ischemia/reperfusion. These effects may serve as a potential novel mechanism of action contributing to the apocynin-induced neuroprotection observed.


Subject(s)
Acetophenones/pharmacology , Apoptosis/drug effects , Brain Ischemia/pathology , Neuroprotective Agents/pharmacology , Reperfusion , Animals , Antioxidants/pharmacology , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Glutathione/analysis , Hydrogen Peroxide/analysis , Male , Middle Cerebral Artery/drug effects , Middle Cerebral Artery/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Stroke/pathology , Stroke/prevention & control
14.
Brain Res ; 1375: 128-36, 2011 Feb 23.
Article in English | MEDLINE | ID: mdl-21172313

ABSTRACT

Lipoic acid (LA) is a known antioxidant currently used as a therapy in patients with vascular and metabolic disorders. We tested the hypothesis that lipoic acid is protective against the cell death observed following stroke. Lipoic acid was administered 30minutes prior to, or immediately following removal of sutures used to occlude the middle cerebral artery (MCA) in male Sprague-Dawley rats. Following removal of the sutures, the MCA territory was allowed to undergo 5.5hrs of reperfusion. This ischemia/reperfusion (I/R) resulted in a focal infarct restricted to the prefrontal cortex (24±3mm(3)). Pretreatment with LA 30minutes prior to occlusion resulted in a dose-dependent reduction in infarct volume. This reduction in infarct volume was not observed when the LA was administered immediately prior to reperfusion (30minutes post-occlusion). To investigate a potential hemodynamic mechanism for this LA-induced neuroprotection, blood pressure, heart rate and baroreceptor reflex sensitivity (BRS) were measured. Intravenous administration of LA did not result in any significant changes in any of these parameters compared to saline-treated rats. Similarly, there was no significant contribution of systemic nitric oxide or alteration in cerebral perfusion measured following pretreatment with lipoic acid or during the course of occlusion and reperfusion compared with saline-treated rats. Western blot analysis of tissue from the ischemic cortex showed an increase in protein expression of superoxide dismutase (SOD2), but not SOD1, in LA pretreated rats. This suggests a potential mechanism of action contributing to the LA-induced neuroprotection observed. Furthermore, the data in the present investigation suggest the potential use of LA pretreatment as a neuroprotectant in stroke patients.


Subject(s)
Antioxidants/pharmacology , Brain Ischemia/complications , Neuroprotective Agents , Reperfusion Injury/prevention & control , Thioctic Acid/pharmacology , Animals , Baroreflex/drug effects , Baroreflex/physiology , Blotting, Western , Brain Ischemia/pathology , Cerebral Infarction/pathology , Cerebrovascular Circulation , Cytosol/enzymology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Enzyme Inhibitors/pharmacology , Hemodynamics/drug effects , Male , Middle Cerebral Artery/drug effects , Middle Cerebral Artery/physiology , Mitochondria/enzymology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/physiology , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
15.
Neurosci Res ; 63(4): 273-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19367787

ABSTRACT

Ischemic tolerance describes a phenomenon whereby subcritical stimuli evoke cellular protective mechanisms resulting in increased tolerance to subsequent ischemia. In the present study we propose that the cytoprotective effects attributed to 17beta-estradiol and tunicamycin in an in vivo rodent model of ischemia are reflected by changes in neuronal tissue levels of m-calpain, HSP70, GRP94 and GRP78. Rats pretreated with 17beta-estradiol, tunicamycin or both demonstrated dose-dependent reductions in infarct area following 4 h of permanent middle cerebral artery occlusion (MCAO). Western blot analysis revealed that 4 h of MCAO was associated with decreased cortical expression of HSP70 and m-calpain and increased expression of GRP78. Pretreatment with 12.5 microg/kg 17beta-estradiol did not change this pattern of protein expression following MCAO. While GRP94 expression was elevated in sham-operated rats pretreated with 17beta-estradiol, the ensuing ischemic tolerance did not appear to be mediated by changes in cellular stress proteins. Pretreatment with 50 microg/kg tunicamycin significantly reduced HSP70 in cortical tissue samples taken from sham-operated rats and appeared to attenuate the threshold for activation of m-calpain in rats undergoing 4 h of MCAO. Lastly, a combined treatment in which rats undergoing MCAO were pretreated with both tunicamycin (24 h prior) and 17beta-estradiol (30 min prior) was associated with an attenuated stress response as indicated by reduced expression of GRP78 and GRP94 when compared to saline-treated controls. The results of this study suggest that the ischemic tolerance observed following MCAO in rats pretreated with either 17beta-estradiol or tunicamycin is likely mediated in part through differential effects on cellular stress proteins.


Subject(s)
Estradiol/pharmacology , Estrogens/pharmacology , Gene Expression Regulation/drug effects , Heat-Shock Proteins/metabolism , Infarction, Middle Cerebral Artery/prevention & control , Animals , Calpain/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , HSP70 Heat-Shock Proteins/metabolism , Infarction, Middle Cerebral Artery/pathology , Male , Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Tunicamycin/pharmacology
16.
Brain Res ; 1247: 212-20, 2009 Jan 09.
Article in English | MEDLINE | ID: mdl-18992720

ABSTRACT

Mild NMDA receptor activation is correlated with neuroprotection in models of cerebral ischemia. Neuroprotection with NMDA manifests as a form of ischemic tolerance and involves the induction of cellular stress systems sensitive to disturbances in cellular calcium homeostasis. Unilateral micro-injection of 10, 160 and 320 microM NMDA into the prefrontal cortex of a rat 30 min prior to permanent occlusion of the middle cerebral artery (MCAO) significantly reduced the area of infarct observed after 4 h of ischemia. The highest dose of NMDA (320 microM) prevented the propagation of ischemic damage through a direct toxicity on neuronal tissue adjacent to the injection site as demonstrated in thionin-stained sections. As a result, the degree of ischemia-induced damage was similar to that measured in rats pretreated with the low dose of NMDA (10 microM). Expression of heat shock protein (HSP) 70 and glucose-regulated protein (GRP) 94 in cortical samples taken from the region of infarct following MCAO was significantly reduced in rats pretreated with 10 microM NMDA compared to saline-injected control rats and rats pretreated with higher doses of NMDA. Furthermore, 10 microM NMDA did not appear to influence expression of m-calpain or GRP78, however, higher doses of NMDA did significantly induce expression of both proteins as assessed by Western blotting. In summary, our data demonstrate an in vivo rodent model of ischemic tolerance in which 30 min of neuronal preconditioning with 10 microM NMDA confers protection against a 4 h period of MCAO-induced ischemia. This effect may involve modulation of cellular stress signals, in particular HSP70 and GRP94.


Subject(s)
Brain/drug effects , HSP70 Heat-Shock Proteins/drug effects , Hypoxia-Ischemia, Brain/drug therapy , Membrane Proteins/drug effects , N-Methylaspartate/pharmacology , Stress, Physiological/drug effects , Animals , Brain/metabolism , Brain/physiopathology , Calpain/drug effects , Calpain/metabolism , Cytoprotection/drug effects , Cytoprotection/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Agonists/therapeutic use , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/drug effects , Heat-Shock Proteins/metabolism , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/physiopathology , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Ischemic Preconditioning/methods , Male , Membrane Proteins/metabolism , Molecular Chaperones/drug effects , Molecular Chaperones/metabolism , N-Methylaspartate/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Rats , Rats, Sprague-Dawley , Stress, Physiological/physiology
17.
Diabetes Metab Res Rev ; 24(5): 392-403, 2008.
Article in English | MEDLINE | ID: mdl-18273840

ABSTRACT

BACKGROUND: Adenosine triphosphate (ATP) is a critical determinant of beta-cell insulin secretion in response to glucose. BHE/cdb rats have a mutation in ATP synthase that limits ATP production, yet develop mild diabetes only with ageing. We investigated the cellular basis for reduced insulin secretion and compensatory mechanisms that mitigate the effects of the ATP synthase mutation. METHODS: In vitro beta-cell function in isolated islets and expression of key regulatory genes was compared with in vivo oral glucose tolerance and insulin sensitivity in BHE/cdb and control rats. RESULTS: BHE/cdb rat islets had reduced responsiveness to glucose stimulation and ATP content was 35% lower than in control islets. Oral glucose tolerance was impaired at both 21 and 43 weeks of age because of a reduction in glucose-stimulated insulin secretion (GSIS). An increase in inducible nitric oxide synthase (INOS, 3-fold) and manganese superoxide dismutase (MnSOD, 1.6-fold), detection of nitrotyrosine, beta-cell apoptosis, and nucleocytoplasmic translocation of pancreas duodenum homeobox-1 (PDX-1) in beta-cells indicated increased oxygen radical formation. However, BHE/cdb rats partially compensated for low glucose responsiveness by increasing the number of small islets and beta-cell hypertrophy. There was also an increase in the proportion of mature insulin relative to proinsulin (PI) detected within beta-cell granules. Increased activation of AMP-dependent kinase (AMPK)-regulated pathways was consistent with increased oxidative stress and with induction of apoptosis and reduction of preproinsulin gene transcription. CONCLUSIONS: The findings are consistent with impaired but partially compensated mechanisms of insulin secretion early in life, but progressive non-compensated impairments due to oxidative stress occurs by age 43 weeks.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Oxidative Stress/physiology , Proton-Translocating ATPases/genetics , Adenosine Triphosphate/metabolism , Animals , Female , Glucose Tolerance Test , Insulin/physiology , Insulin Secretion , Insulin-Secreting Cells/cytology , Islets of Langerhans/metabolism , Phenotype , Phosphotransferases (Phosphate Group Acceptor)/physiology , Proinsulin/metabolism , Rats
18.
J Endocrinol ; 190(3): 659-67, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17003267

ABSTRACT

We hypothesized that the loss of glucose homeostasis in ob/ob mice is associated with upregulation of islet uncoupling protein-2 (UCP2) expression, leading to impaired glucose-stimulated insulin secretion (GSIS). Changes in glucose homeostasis in lean and ob/ob mice from 5 to 16 weeks were assessed by fasting blood glucose, plasma insulin, oral glucose tolerance, and tissue insulin sensitivity. In vitro GSIS and ATP content were assayed in isolated islets, while UCP2 expression was determined by quantitative real-time PCR and immunoblotting. Short-term reduction of UCP2 expression was achieved through transfection of islets with specific small interfering RNA. Insulin resistance was detected in 5-week-old ob/ob mice, but GSIS and blood glucose levels remained normal. By 8 weeks of age, ob/ob mice displayed fasting hyperglycemia, hyperinsulinemia and glucose intolerance, and also had elevated non-esterified fatty acid concentration in plasma. In vitro, GSIS and ATP generation were impaired in ob/ob islets. Islet UCP2 expression was elevated at 5 and 8 weeks of age. Short-term knockdown of islet UCP2 increased GSIS in islets of lean mice, but had no effect in islets from ob/ob mice. Loss of glucose homeostasis and impairment of insulin secretion from isolated islets at 8 weeks in ob/ob mice is preceded by an increase in UCP2 expression in islets. Moreover, the glucolipotoxic conditions observed are predicted to increase UCP2 activity, contributing to lower islet ATP and GSIS.


Subject(s)
Glucose/metabolism , Insulin-Secreting Cells/metabolism , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Obesity/metabolism , Up-Regulation , Adenosine Triphosphate/analysis , Adenosine Triphosphate/metabolism , Aging/physiology , Animals , Blood Glucose/analysis , Cells, Cultured , Female , Glucose Tolerance Test , Homeostasis , Immunoblotting , Insulin/blood , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Ion Channels/analysis , Ion Channels/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mitochondrial Proteins/analysis , Mitochondrial Proteins/genetics , PPAR gamma/analysis , PPAR gamma/genetics , RNA, Messenger/analysis , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Uncoupling Protein 2
19.
J Biol Chem ; 279(49): 51049-56, 2004 Dec 03.
Article in English | MEDLINE | ID: mdl-15448158

ABSTRACT

Chronic exposure to elevated free fatty acids (lipotoxicity) induces uncoupling protein (UCP2) in the pancreatic beta-cell, and therefore a causal link between UCP2 and beta-cell defects associated with obesity may exist. Recently, we showed that lipid treatment in vivo and in vitro in UCP2(-/-) mice/islets does not result in any loss in beta-cell glucose sensitivity. We have now assessed the mechanism of maintained beta-cell function in UCP2(-/-) mice by exposing islets to 0.4 mM palmitate for 48 h. Palmitate treatment increased triglyceride concentrations in wild type (WT) but not UCP2(-/-) islets because of higher palmitate oxidation rates in the UCP2(-/-) islets. Dispersed beta-cells from the palmitate-exposed WT islets had reduced glucose-stimulated hyperpolarization of the mitochondrial membrane potential compared with both control WT and palmitate-exposed UCP2(-/-) beta-cells. The glucose-stimulated increases in the ATP/ADP ratio and cytosolic Ca2+ are attenuated in palmitate-treated WT but not UCP2(-/-) beta-cells. Exposure to palmitate reduced glucose-stimulated insulin secretion (GSIS) in WT islets, whereas UCP2(-/-) islets had enhanced GSIS. Overexpression of recombinant UCP2 but not enhanced green fluorescent protein in beta-cells resulted in a loss of glucose-stimulated hyperpolarization of the mitochondrial membrane potential and GSIS similar to that seen in WT islets exposed to palmitate. Reactive oxygen species (ROS) are known to increase the activity of UCP2. We showed that ROS levels were elevated in control UCP2(-/-) islets as compared with WT and UCP2(-/-) islets overexpressing UCP2 and that palmitate increased ROS in WT and UCP2(-/-) islets overexpressing UCP2 but not in UCP2(-/-) islets. Thus, UCP2(-/-) islets resisted the toxic effects of palmitate by maintaining glucose-dependent metabolism-secretion coupling. We propose that higher free fatty acid oxidation rates prevent accumulation of triglyceride in UCP2(-/-) islets, such accumulation being a phenomenon associated with lipotoxicity.


Subject(s)
Fatty Acids, Nonesterified/metabolism , Islets of Langerhans/metabolism , Membrane Transport Proteins/biosynthesis , Mitochondrial Proteins/biosynthesis , Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Adenoviridae/genetics , Animals , Blotting, Western , Calcium/chemistry , Calcium/metabolism , Cytosol/metabolism , Dose-Response Relationship, Drug , Fatty Acids/metabolism , Glucose/chemistry , Glucose/metabolism , Insulin/blood , Insulin/metabolism , Ion Channels , Lipid Metabolism , Male , Membrane Potentials , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Obesity/metabolism , Oxygen/metabolism , Palmitic Acid/metabolism , Phenotype , Polymorphism, Genetic , Reactive Oxygen Species , Recombinant Proteins/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transgenes , Triglycerides/chemistry , Triglycerides/metabolism , Uncoupling Protein 2
20.
Diabetes ; 53 Suppl 1: S136-42, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14749279

ABSTRACT

Stressors such as chronic hyperglycemia or hyperlipidemia may lead to insufficient insulin secretion in susceptible individuals, contributing to type 2 diabetes. The molecules mediating this effect are just beginning to be identified. Uncoupling protein (UCP)-2 may be one such negative modulator of insulin secretion. Accumulating evidence shows that beta-cell UCP2 expression is upregulated by glucolipotoxic conditions and that increased activity of UCP2 decreases insulin secretion. Mitochondrial superoxide has been identified as a posttranslational regulator of UCP2 activity in islets; thus, UCP2 may provide protection to beta-cells at one level while simultaneously having detrimental effects on insulin secretion. Interestingly, the latter appears to be the dominant outcome, because UCP2 knockout mice display an increased beta-cell mass and retained insulin secretion capacity in the face of glucolipotoxicity.


Subject(s)
Islets of Langerhans/physiology , Membrane Transport Proteins/physiology , Mitochondrial Proteins/physiology , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Gene Expression Regulation/physiology , Humans , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Ion Channels , Nutritional Physiological Phenomena , Reactive Oxygen Species/metabolism , Uncoupling Protein 2
SELECTION OF CITATIONS
SEARCH DETAIL
...