Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Vet Surg ; 51(5): 781-787, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35500144

ABSTRACT

OBJECTIVE: To determine the influence of the staple line configuration on the leakage of small intestinal functional end-to-end stapled anastomosis (FEESA). STUDY DESIGN: Experimental, ex vivo, randomized study. SAMPLE POPULATION: Jejunal segments (N = 72) from 10 mature, canine cadavers. METHODS: Jejunal segments (10 cm) were randomly assigned to a control group (8 segments) and 4 FEESA groups (16 segments/group (8 constructs/group)), according to the number of rows of staples used in the vertical (V) and transverse lines (T), respectively: Control, 2-row V/2-row T (2V/2T), 2-row V/3-row T (2V/3T), 3-row V/2-row T (3V/2T), 3-row V/3-row T (3V/3T). Initial leak pressure (ILP), maximum intraluminal pressure (MIP), and initial leakage location (ILL) were compared. RESULTS: The ILP (mean ± SD) for control segments, 2V/2T, 2V/3T, 3V/2T and 3V/3T were 321.38 ± 34.59, 32.88 ± 7.36, 50.13 ± 10.46, 34.38 ± 11.78, 69.88 ± 21.23 mmHg, respectively. All FEESAs initially leaked at lower pressures than intact segments. The only other differences detected between groups consisted of ILPs that were higher when FEESAs were closed with 3V/3T (69.88 ± 21.23 mmHg) than 2V/2T (32.88 ± 7.36, P < .001). Initial leakage occurred predominantly from the transverse staple line rather than the anastomotic crotch (P < .001). CONCLUSION: Placing 3 rows of staples in the transverse line (with or without a third row in the vertical staple line) improved resistance to leakage of FEESAs in normal cadaveric specimens. CLINICAL SIGNIFICANCE: The addition of a third row of staples in the transverse line (with or without a third row in the vertical staple line) in FEESAs should be further investigated as a strategy to reduce intestinal leakage clinically.


Subject(s)
Intestine, Small , Sutures , Anastomosis, Surgical/veterinary , Animals , Dogs , Intestine, Small/surgery , Pressure , Surgical Stapling/veterinary
2.
Antioxidants (Basel) ; 11(2)2022 Jan 23.
Article in English | MEDLINE | ID: mdl-35204096

ABSTRACT

It is estimated that the prevalence rate of Alzheimer's disease (AD) will double by the year 2040. Although currently available treatments help with symptom management, they do not prevent, delay the progression of, or cure the disease. Interestingly, a shared characteristic of AD and other neurodegenerative diseases and disorders is oxidative stress. Despite profound evidence supporting the role of oxidative stress in the pathogenesis and progression of AD, none of the currently available treatment options address oxidative stress. Recently, attention has been placed on the use of antioxidants to mitigate the effects of oxidative stress in the central nervous system. In preclinical studies utilizing cellular and animal models, natural antioxidants showed therapeutic promise when administered alone or in combination with other compounds. More recently, the concept of combination antioxidant therapy has been explored as a novel approach to preventing and treating neurodegenerative conditions that present with oxidative stress as a contributing factor. In this review, the relationship between oxidative stress and AD pathology and the neuroprotective role of natural antioxidants from natural sources are discussed. Additionally, the therapeutic potential of natural antioxidants as preventatives and/or treatment for AD is examined, with special attention paid to natural antioxidant combinations and conjugates that are currently being investigated in human clinical trials.

3.
Int J Mol Sci ; 24(1)2022 Dec 27.
Article in English | MEDLINE | ID: mdl-36613883

ABSTRACT

Antioxidants are being explored as novel therapeutics for the treatment of neurodegenerative diseases such as Alzheimer's disease (AD) through strategies such as chemically linking antioxidants to synthesize novel co-drugs. The main objective of this study was to assess the cytoprotective effects of the novel antioxidant compound VANL-100 in a cellular model of beta-amyloid (Aß)-induced toxicity. The cytotoxic effects of Aß in the presence and absence of all antioxidant compounds were measured using the 3-(4,5-dimethylthiazol-2-yl)2-5-diphenyl-2H-tetrazolium bromide (MTT) assay in SH-SY5Y cells in both pre-treatment and co-treatment experiments. In pre-treatment experiments, VANL-100, or one of its parent compounds, naringenin (NAR), alpha-lipoic acid (ALA), or naringenin + alpha-lipoic acid (NAR + ALA), was administrated 24 h prior to an additional 24-h incubation with 20 µM non-fibril or fibril Aß25-35. Co-treatment experiments consisted of simultaneous treatment with Aß and antioxidants. Pre-treatment and co-treatment with VANL-100 significantly attenuated Aß-induced cell death. There were no significant differences between the protective effects of VANL-100, NAR, ALA, and NAR + ALA with either form of Aß, or in the effect of VANL-100 between 24-h pre-treatment and co-treatment. These results demonstrate that the novel co-drug VANL-100 is capable of eliciting cytoprotective effects against Aß-induced toxicity.


Subject(s)
Alzheimer Disease , Antioxidants , Neuroprotective Agents , Thioctic Acid , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/toxicity , Antioxidants/pharmacology , Cell Line, Tumor , Neuroprotective Agents/pharmacology , Peptide Fragments/toxicity , Thioctic Acid/pharmacology
4.
Vet Surg ; 49 Suppl 1: O120-O130, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32053219

ABSTRACT

OBJECTIVE: To evaluate the performance of an endoscopic 3-mm electrothermal bipolar vessel sealing device (EBVS) intended for single use after multiple use-and-resterilization cycles. STUDY DESIGN: Ex vivo study. SAMPLE POPULATION: Eight 3-mm EBVS handpieces. METHODS: Handpieces were subjected to a maximum of 15 cycles of testing, including simulated surgery, sealing and burst pressure testing of porcine carotid arteries, reprocessing, and hydrogen peroxide plasma resterilization. Failure was defined as two sequential vascular seal leakage events occurring at <250 mm Hg. Histological evaluation, maximum external temperature of the jaws, sealing time, tissue adherence, jaw surface characterization, and mechanical deterioration were studied. Failure rate was analyzed by using a Kaplan-Meier curve. Linear and ordinal logistic mixed models were used to analyze sealing time, handpiece jaw temperature, and adherence score. RESULTS: Mean ± SD diameter of arteries was 3.22 ± 0.35 mm. Failure was observed starting at cycle 10 and going up to cycle 13 in 37.5% (3/8) of the handpieces. Tissue adherence increased after each cycle (P < .001). Maximum external temperature (79.8°C ± 13.9°C) and sealing time (1.8 ± 0.5 seconds) were not significantly different throughout cycles up to failure. A flatter surface and large scratches were observed microscopically throughout the jaw surface after repeated use and resterilization. CONCLUSION: The 3-mm EBVS handpiece evaluated in this study can be considered safe to use for up to nine reuse-and-resterilization cycles. CLINICAL SIGNIFICANCE: These data provide the basis for establishing preliminary guidelines for the reuse and hydrogen peroxide plasma resterilization of an endoscopic 3-mm EBVS handpiece.


Subject(s)
Electrocoagulation/veterinary , Sterilization , Surgical Instruments/veterinary , Vascular Surgical Procedures/instrumentation , Animals , Carotid Arteries , Electrocoagulation/instrumentation , Swine
5.
Int J Artif Organs ; 41(8): 421-430, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29807488

ABSTRACT

Decellularization of tissues can significantly improve regenerative medicine and tissue engineering by producing natural, less immunogenic, three-dimensional, acellular matrices with high biological activity for transplantation. Decellularized matrices retain specific critical components of native tissues such as stem cell niche, various growth factors, and the ability to regenerate in vivo. However, recellularization and functionalization of these matrices remain limited, highlighting the need to improve the characteristics of decellularized matrices. Incorporating nanoparticles into decellularized tissues can overcome these limitations because nanoparticles possess unique properties such as multifunctionality and can modify the surface of decellularized matrices with additional growth factors, which can be loaded onto the nanoparticles. Therefore, in this minireview, we highlight the various approaches used to improve decellularized matrices with incorporation of nanoparticles and the challenges present in these applications.


Subject(s)
Extracellular Matrix/chemistry , Nanoparticles , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Humans , Regeneration , Regenerative Medicine/methods
6.
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
7.
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
8.
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
9.
Neurosci Lett ; 606: 117-22, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26320021

ABSTRACT

Inflammation-related cerebral damage mediated by infiltrating neutrophils following reperfusion plays a role in reperfusion-induced brain damage subsequent to a stroke event. The ELR-CXC family of chemokines are CXCR1 and CXCR2 agonists that are known to drive neutrophil migration and activation. The present study demonstrated the benefit of anti-inflammatory therapy in the treatment of ischemic stroke with the administration of the competitive ELR-CXC chemokine antagonist, CXCL8(3-72)K11R/G31P (G31P). Male Sprague-Dawley rats were anaesthetized, and the middle cerebral artery (MCA) was occluded for 30 min followed by 5.5 h of reperfusion. Pretreatment with G31P resulted in a significant, dose-dependent (approximately 61-72%) decrease in infarct volumes compared to vehicle-treated animals, but neuroprotection was also observed when G31P (0.5 mg/kg) was administered 1 or 3 h following the start of reperfusion. The neuroprotection observed following the administration of this competitive CXCR1/CXCR2 antagonist may present therapeutic opportunities for addressing reperfusion-induced inflammatory damage in patients presenting with transient ischemic episodes.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Brain Ischemia/drug therapy , Chemokines, CXC/antagonists & inhibitors , Interleukin-8/therapeutic use , Neuroprotective Agents/therapeutic use , Peptide Fragments/therapeutic use , Stroke/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Brain/blood supply , Brain/drug effects , Brain/pathology , Brain Infarction/drug therapy , Brain Infarction/pathology , Brain Ischemia/etiology , Brain Ischemia/immunology , Brain Ischemia/pathology , Chemokines, CXC/immunology , Infarction, Middle Cerebral Artery/complications , Interleukin-8/pharmacology , Male , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Rats, Sprague-Dawley , Stroke/etiology , Stroke/immunology , Stroke/pathology
10.
Brain Sci ; 5(2): 130-43, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25906324

ABSTRACT

Previous work in our laboratory demonstrated the utility of synthetic combinations of two naturally occurring, biologically active compounds. In particular, we combined two known anti-oxidant compounds, lipoic acid and apocynin, covalently linked via an ester bond (named UPEI-100). In an animal model of ischemia-reperfusion injury (tMCAO), UPEI-100 was shown to produce equivalent neuroprotection compared to each parent compound, but at a 100-fold lower dose. However, it was determined that UPEI-100 was undetectable in any tissue samples almost immediately following intravenous injection. Therefore, the present investigation was done to determine if biological stability of UPEI-100 could be improved by replacing the ester bond with a more bio cleavage-resistant bond, an ether bond (named UPEI-104). We then compared the stability of UPEI-104 to the original parent compound UPEI-100 in human plasma as well as liver microsomes. Our results demonstrated that both UPEI-100 and UPEI-104 could be detected in human plasma for over 120 min; however, only UPEI-104 was detectable for an average of 7 min following incubation with human liver microsomes. This increased stability did not affect the biological activity of UPEI-104 as measured using our tMCAO model. Our results suggest that combining compounds using an ether bond can improve stability while maintaining biological activity.

11.
Endocrinology ; 156(2): 638-46, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25456068

ABSTRACT

The pregnancy hormone relaxin protects tissue from ischemic damage. The ability of relaxin-3, a relaxin paralog, to do so has not been explored. The cerebral expression levels of these peptides and their receptors make them logical targets for study in the ischemic brain. We assessed relaxin peptide-mediated protection, relative relaxin family peptide receptor (RXFP) involvement, and protective mechanisms. Sprague-Dawley rats receiving permanent (pMCAO) or transient middle cerebral artery occlusions (tMCAO) were treated with relaxin peptides, and brains were collected for infarct analysis. Activation of the endothelial nitric oxide synthase pathway was evaluated as a potential protective mechanism. Primary cortical rat astrocytes were exposed to oxygen glucose deprivation and treated with relaxin peptides, and viability was examined. Receptor involvement was explored using RXFP3 antagonist or agonist treatment and real-time PCR. Relaxin and relaxin-3 reduced infarct size after pMCAO. Both peptides activated endothelial nitric oxide synthase. Because relaxin-3 has not previously been associated with this pathway and displays promiscuous RXFP binding, we explored the receptor contribution. Expression of rxfp1 was greater than that of rxfp3 in rat brain, although peptide binding at either receptor resulted in similar overall protection after pMCAO. Only RXFP3 activation reduced infarct size after tMCAO. In astrocytes, rxfp3 gene expression was greater than that of rxfp1. Selective activation of RXFP3 maintained astrocyte viability after oxygen glucose deprivation. Relaxin peptides are protective during the early stages of ischemic stroke. Differential responses among treatments and models suggest that RXFP1 and RXFP3 initiate different protective mechanisms. This preliminary work is a pivotal first step in identifying the clinical implications of relaxin peptides in ischemic stroke.


Subject(s)
Infarction, Middle Cerebral Artery/prevention & control , Relaxin/therapeutic use , Animals , Astrocytes/drug effects , Brain/pathology , Cells, Cultured , Disease Models, Animal , Drug Evaluation, Preclinical , Infarction, Middle Cerebral Artery/pathology , Male , Random Allocation , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Peptide/agonists , Receptors, Peptide/antagonists & inhibitors , Recombinant Proteins/therapeutic use , Relaxin/pharmacology
12.
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
13.
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
14.
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
15.
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
16.
Cardiol Res Pract ; 2012: 319230, 2012.
Article in English | MEDLINE | ID: mdl-22263115

ABSTRACT

Background. Lipoic acid (LA), which has significant antioxidant properties, may also function as a potent neuroprotectant. The synthetic compounds INV-155, INV-157, INV-159, and INV-161 are physiochemical combinations of lipoic acid and captopril. We sought to determine if these compounds have neuroprotective potential following middle cerebral artery occlusion (MCAO) in rats. Methods. Male Sprague-Dawley rats were injected intravenously with captopril (1-50 mg/kg) 30 minutes prior to MCAO. Blood pressure, heart rate, baroreceptor reflex sensitivity, and infarct size were measured. In addition, dose response effect on infarct size and cardiovascular parameters was determined using INV-155, INV-157, INV-159, and INV-161 and compared to captopril and LA. Results. Pretreatment with captopril and LA at all doses tested was neuroprotective. The compounds INV-159 (0.5-10 mg/kg) and INV-161 (1-10 mg/kg) produced a significant,dose-dependent decrease in infarct size. In contrast, INV-155 and INV-157 had no effect on infarct size. Conclusions. Combined pretreatment with captopril potentiated the neuroprotective benefit observed following LA alone. Both INV-159 and INV-161 were also neuroprotective. These results suggest that patients taking combinations of captopril and LA, either as combination therapy or in the form of INV-159 or INV-161, may also benefit from significant protection against cerebral infarction.

17.
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
18.
Neuropharmacology ; 62(2): 825-32, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21930135

ABSTRACT

Substance P (SP) and cocaine employ similar mechanisms to modify excitatory synaptic transmission in the nucleus accumbens (NAc), a region implicated in substance abuse. Here we explored, using NAc slices, whether SP effects on these synaptic responses were altered in rats that have been sensitized to cocaine and whether SP could mimic cocaine in triggering increased locomotion in sensitized rats. Intraperitoneal (IP) injection of naïve rats with cocaine (15 mg/kg) caused increased locomotion by 408.5 ± 85.9% (n = 5) which further increased by 733.1 ± 157.8% (n = 5) following a week of cocaine sensitization. A similar challenge with 10 mg/kg of SP after cocaine sensitization did not produce significant changes in locomotion (170.6 ± 61.0%; n = 4). In contrast to cocaine, IP injection of rats with SP or SP(5-11) (10-100 mg/kg) with or without phosphoramidon did not elicit changes in locomotion. In electrophysiological studies, both cocaine and SP depressed evoked NMDA and non-NMDA receptor-mediated excitatory synaptic currents (EPSCs) in slices obtained from naïve rats. In slices derived from cocaine-sensitized rats, cocaine but not SP produced a more profound decrease in non-NMDA compared to NMDA responses. Similar to that in naïve rats, cocaine's effect on the EPSCs in these sensitized rats occluded those of SP. Thus, although SP and cocaine may employ similar mechanisms to depress EPSCs in the NAc, IP injection of SP does not mimic cocaine-induced hyperlocomotion indicating that not all of cocaine's effects are mimicked by SP. This article is part of a Special Issue entitled 'Post-Traumatic Stress Disorder'.


Subject(s)
Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Motor Activity/drug effects , Nucleus Accumbens/drug effects , Substance P/pharmacology , Synaptic Transmission/drug effects , Animals , Locomotion/drug effects , Male , Rats , Rats, Sprague-Dawley
19.
Neurosci Lett ; 507(1): 43-6, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22155048

ABSTRACT

Apocynin is a well-known NADPH-oxidase inhibitor currently being investigated for its potential therapeutic use in patients with cardiovascular disease, such as occlusive stroke. However, the use of apocynin as a potential neuroprotective agent has come under criticism due to a narrow experimental therapeutic dose range and possible pro-oxidant effects at high doses. Lipoic acid is a powerful antioxidant due to its ability to scavenge free radicals at very low doses and has been demonstrated to enhance the therapeutic value of several other classes of drugs. Therefore, the present study was designed to determine if co-administration of previously determined non-neuroprotective doses of lipoic acid and apocynin in combination could enhance their neuroprotective ability thus extending the therapeutic dose range. We tested the hypothesis in a rat model of stroke and reperfusion injury. The middle cerebral artery (MCA) in male Sprague-Dawley rats was occluded for 30 min followed by 5.5h of reperfusion. Pre-treatment with several doses of apocynin (0.05, 0.1 and 1.0 mg/kg) in combination with a single dose of lipoic acid (0.005 mg/kg) resulted in a dose-dependent reduction in infarct volume up to ~50%. These results demonstrate that a non-effective dose of lipoic acid can enhance the neuroprotective ability of apocynin at doses which were previously demonstrated to be non-neuroprotective. Co-administration of apocynin with lipoic acid may overcome the criticisms of the use of apocynin as a neuroprotectant and provide an effective therapy in the prevention of cell death following stroke.


Subject(s)
Acetophenones/administration & dosage , Brain Ischemia/drug therapy , Brain Ischemia/physiopathology , Brain/physiopathology , Reperfusion Injury/drug therapy , Reperfusion Injury/physiopathology , Thioctic Acid/administration & dosage , Animals , Brain/drug effects , Drug Combinations , Humans , Male , Neuroprotective Agents , Rats , Rats, Sprague-Dawley , Treatment Outcome
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL