Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Neural Regen Res ; 14(12): 2192-2198, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31397359

ABSTRACT

Spinal cord ischemia associated with trauma and surgical procedures including thoraco-abdominal aortic aneurysm repair and thoracic endovascular aortic repair results in devastating clinical deficits in patients. Because spinal cord ischemia is inadequately treated, we studied the effects of [4-((1E)-2-(5-(4-hydroxy-3-methoxystyryl-)-1-phenyl-1H-pyrazoyl-3-yl) vinyl)-2-methoxy-phenol)] (CNB-001), a novel curcumin-based compound, in a rabbit SCI model. CNB-001 is known to inhibit human 5-lipoxygenase and 15-lipoxygenase and reduce the ischemia-induced inflammatory response. Moreover, CNB-001 can reduce the level of oxidative stress markers and potentiate brain-derived neurotrophic factor and brain-derived neurotrophic factor receptor signaling. The Tarlov scale and quantal analysis technique results revealed that CNB-001 administered as an intravenous dose (bolus) 30 minutes prior to spinal cord ischemia improved the behaviors of female New Zealand White rabbits. The improvements were similar to those produced by the uncompetitive N-methyl-D-aspartate receptor antagonist memantine. At 48 hours after aortic occlusion, there was a 42.7% increase (P < 0.05) in tolerated ischemia duration (n = 14) for rabbits treated with CNB-001 (n = 16), and a 72.3% increase for rabbits treated with the positive control memantine (P < 0.05) (n = 23) compared to vehicle-treated ischemic rabbits (n = 22). CNB-001 is a potential important novel treatment for spinal cord ischemia induced by aortic occlusion. All experiments were approved by the CSMC Institutional Animal Care and Use Committee (IACUC #4311) on November 1, 2012.

2.
Exp Neurol ; 313: 98-108, 2019 03.
Article in English | MEDLINE | ID: mdl-30521790

ABSTRACT

Ischemic stroke is an acute neurodegenerative disease that is extremely devastating to patients, their families and society. Stroke is inadequately treated even with endovascular procedures and reperfusion therapy. Using an extensive translational screening process, we have developed a pleiotropic cytoprotective agent with the potential to positively impact a large population of brain ischemia patients and revolutionize the process used for the development of new drugs to treat complex brain disorders. In this unique translational study article, we document that the novel curcumin-based compound, CNB-001, when administered as a single intravenous dose, has significant efficacy to attenuate clinically relevant behavioral deficits following ischemic events in agyrencephalic rabbits when administered 1 h post-embolization and reduces infarct growth in gyrencephalic non-human primates, when administered 5 min after initiation of middle cerebral artery occlusion. CNB-001 is safe and does not increase morbidity or mortality in either research species. Mechanistically, CNB-001 inhibits human 5- and 15-lipoxygenase in vitro, and can attenuate ischemia-induced inflammatory markers, and oxidative stress markers, while potentially promoting synaptic plasticity mediated by enhanced brain-derived neurotrophic factor (BDNF).


Subject(s)
Brain Ischemia/drug therapy , Curcumin/analogs & derivatives , Lipoxygenase Inhibitors/therapeutic use , Neuroprotective Agents/therapeutic use , Pyrazoles/therapeutic use , Stroke/drug therapy , Administration, Intravenous , Animals , Behavior, Animal/drug effects , Brain Ischemia/psychology , Curcumin/pharmacokinetics , Curcumin/pharmacology , Curcumin/therapeutic use , Disease Progression , Infarction, Middle Cerebral Artery/drug therapy , Lipoxygenase Inhibitors/pharmacokinetics , Lipoxygenase Inhibitors/pharmacology , Macaca fascicularis , Magnetic Resonance Imaging , Male , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Rabbits , Stroke/psychology
3.
Exp Neurol ; 307: 109-117, 2018 09.
Article in English | MEDLINE | ID: mdl-29908146

ABSTRACT

Acute ischemic stroke is devastating to patients and their families because of few viable therapeutic options to promote recovery after reperfusion windows close. Recent breakthroughs in biotechnology have resulted in a reproducible patented process for the purification of extracellular vesicles (EVs) from human cardiosphere-derived cells (CDCs). Because CDC-EVs have many features potentially beneficial to treat acute ischemic stroke, CDC-EVs were evaluated in an established small-clot rabbit embolic stroke model, where clinically relevant end points were used to assess recovery in a more translational large animal model. Biodistribution studies with fluorescent DiD-labeled CDC-EVs showed intense uptake in the ischemic region of the brain. In this report, we show that intravenous (IV) CDC-EVs (0.75 mg/kg) administered 1-hour post-embolization significantly attenuate behavioral deficits following an embolic stroke in rabbits. In CDC-EV-treated rabbits, P50 (3.63 ±â€¯1.27 mg, n = 24) was increased by 245% over vehicle control (1.05 ±â€¯0.15 mg, n = 24); by comparison, rt-PA increased P50 by 91% (2.01 ±â€¯0.24 mg, n = 23). Importantly, the therapy was also without adverse effects on intracerebral hemorrhage or survival rate of embolized rabbits. Thus, as a first step toward widespread use, CDC-EVs, given adjunctively to routine reperfusion therapy, merit further investigation as a therapeutic candidate for stroke.


Subject(s)
Exosomes/transplantation , Extracellular Vesicles/transplantation , Recovery of Function , Stroke/therapy , Thromboembolism/therapy , Thrombosis/therapy , Administration, Intravenous , Animals , Exosomes/physiology , Extracellular Vesicles/physiology , Humans , Male , Pilot Projects , Proof of Concept Study , Rabbits , Random Allocation , Recovery of Function/physiology , Stroke/pathology , Thromboembolism/pathology , Thrombosis/pathology
5.
Transl Stroke Res ; 9(1): 4-8, 2018 02.
Article in English | MEDLINE | ID: mdl-28283966

ABSTRACT

Important questions regarding the conduct of scientific research and data transparency have been raised in various scientific forums over the last 10 years. It is becoming clear, that in spite of published RIGOR guidelines, that improvement in the transparency of scientific research is required to focus on the discovery and drug development process so that a treatment can be provided to stroke patients. We have the unique privilege of conducting research using animal models of a disease so that we can address the development of a new therapy, and we should do this with great care and vigilance. This document identifies valuable resources for researchers to become Good Laboratory Practices compliant and increase and improve data transparency and provides guidelines for accurate data management to continue to propel the translational stroke research field forward while recognizing that there is a shortage of research funds worldwide. While data audits are being considered worldwide by funding agencies and they are used extensively by industry, they are still quite controversial for basic researchers. Due to the special exploratory nature of basic and translational science research, the current challenging funding environment, and independent and individualized laboratory activities, it is debatable if current individualized non-standardized data management and monitoring represents the best approach. Thus, herein, we propose steps to prepare research study data in an acceptable form for archival purposes so that standards for translational research data can be comparable to those that are accepted and adhered to by the clinical community. If all translational research laboratories follow and institute the guidelines while conducting translational research, data from all sources may be more comparable and reliable.


Subject(s)
Clinical Trials as Topic/methods , Clinical Trials as Topic/standards , Data Accuracy , Stroke/therapy , Translational Research, Biomedical/standards , Animals , Humans
6.
Transl Stroke Res ; 2017 Apr 12.
Article in English | MEDLINE | ID: mdl-28405804

ABSTRACT

Tissue plasminogen activator (tPA) is currently used in combination with endovascular procedures to enhance recanalization and cerebral reperfusion and is also currently administered as standard-of-care thrombolytic therapy to patients within 3-4.5 h of an ischemic stroke. Since tPA is not neuroprotective or cytoprotective, adjuvant therapy with a neuroprotective or an optimized cytoprotective compound is required to provide the best care to stroke victims to maximally promote clinical recovery. In this article, we describe the use of a sensitive standardized protease assay with CH3SO2-D-hexahydrotyrosine-Gly-Arg-p-nitroanilide•AcOH, a chromogenic protease substrate that is cleaved to 4-nitroaniline (p-nitroaniline) and measured spectrophotometrically at 405 nm (OD405 nm), and how the assay can be used as an effective screening assay to study drug-tPA interactions. While we focus on two compounds of interest in our drug development pipeline, the assay is broadly applicable to all small molecule neuroprotective or cytoprotective compounds currently being discovered and developed worldwide. In this present study, we found that the specific tPA inhibitor, plasminogen activator inhibitor-1 (PAI-1; 0.25 µM), significantly (p < 0.0001) inhibited 4-nitroaniline release, by 97.74% during the 10-min duration of the assay, which is indicative of tPA protease inhibition. In addition, two lead chromone cytoprotective candidates, 2-(3',4',5'-trihydroxyphenyl)chromen-4-one (3',4',5'-trihydroxyflavone) (CSMC-19) and 3-hydroxy-2-[3-hydroxy-4-(pyrrolidin-1-yl)phenyl]benzo[h]chromen-4-one (CSMC-140), also significantly (p < 0.05) reduced 4-nitroaniline accumulation, but to a lesser extent. The reduction was 68 and 45%, respectively, at 10 µM, and extrapolated IC50 values were 4.37 and >10 µM for CSMC-19 and CSMC-140, respectively. Using bonafide 4-nitroaniline, we then demonstrated that the reduction of 4-nitroaniline detection was not due to drug-4-nitroaniline quenching of signal detection at OD405 nm. In conclusion, the results suggest that high concentrations of both cytoprotectives reduced 4-nitroaniline production in vitro, but the inhibition only occurs with concentrations 104-1025-fold that of EC50 values in an efficacy assay. Thus, CSMC-19 and CSMC-140 should be further developed and evaluated in embolic stroke models in the absence or presence of a thrombolytic. If necessary, they could be administered once effective tPA thrombolysis has been confirmed to avoid the possibility that the chromone will reduce the efficacy of tPA in patients. Stroke investigator developing new cytoprotective small molecules should consider adding this sensitive assay to their development and screening repertoire to assess possible drug-tPA interactions in vitro as a de-risking step.

9.
Transl Stroke Res ; 8(4): 307-317, 2017 08.
Article in English | MEDLINE | ID: mdl-28039575

ABSTRACT

Acute ischemic stroke is inadequately treated in the USA and worldwide due to a lengthy history of neuroprotective drug failures in clinical trials. The majority of victims must endure life-long disabilities that not only affect their livelihood, but also have an enormous societal economic impact. The rapid development of a neuroprotective or cytoprotective compound would allow future stroke victims to receive a treatment to reduce disabilities and further promote recovery of function. This opinion article reviews in detail the enormous costs associated with developing a small molecule to treat stroke, as well as providing a timely overview of the cell-death time-course and relationship to the ischemic cascade. Distinct temporal patterns of cell-death of neurovascular unit components provide opportunities to intervene and optimize new cytoprotective strategies. However, adequate research funding is mandatory to allow stroke researchers to develop and test their novel therapeutic approach to treat stroke victims.


Subject(s)
Cytoprotection , Neuroprotective Agents/economics , Neuroprotective Agents/therapeutic use , Stroke/drug therapy , Stroke/economics , Animals , Cost-Benefit Analysis , Humans , Thrombolytic Therapy , Translational Research, Biomedical
10.
Brain Res ; 1646: 125-131, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27180104

ABSTRACT

Tissue plasminogen activator (tPA) is the only FDA-approved treatment for stroke; tPA increases cerebral reperfusion, blood flow and improved behavior. Novel transcranial laser therapy (TLT) also enhances cerebral blood flow and activates mitochondrial function. Using the rabbit small clot embolic stroke model (RSCEM), we studied the effects of continuous wave TLT (7.5mW/cm(2)) alone or in combination with standardized intravenous (IV) tPA (3.3mg/kg) applied 1h post-embolization on 3 endpoints: 1) behavioral function measured 2 days [effective stroke dose (P50 in mg) producing neurological deficits in 50% of embolized rabbits], 2) intracerebral hemorrhage (ICH) rate, and 3) cortical adenosine-5'-triphosphate (ATP) content was measured 6h following embolization. TLT and tPA significantly (p<0.05) increased P50 values by 95% and 56% (p<0.05), respectively over control. TLT-tPA increased P50 by 136% over control (p<0.05). Embolization reduced cortical ATP content by 39%; decreases that were attenuated by either TLT or tPA treatment (p<0.05). TLT-tPA further enhanced cortical ATP levels 22% above that measured in naïve control. TLT and tPA both effectively and safely, without affecting ICH rate, improved behavioral outcome in embolized rabbits; and there was a trend (p>0.05) for the TLT-tPA combination to further increase P50. TLT and tPA both attenuated stroke-induced ATP deficits, and the combination of tPA and TLT produced an additive effect on ATP levels. This study demonstrates that the combination of TLT-tPA enhances ATP production, and suggests that tPA-induced reperfusion in combination with TLT neuroprotection therapy may optimally protect viable cells in the cortex measured using ATP levels as a marker.


Subject(s)
Behavior, Animal/drug effects , Fibrinolytic Agents/administration & dosage , Laser Therapy , Mitochondria/drug effects , Stroke/therapy , Tissue Plasminogen Activator/administration & dosage , Adenosine Triphosphate/metabolism , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Fibrinolytic Agents/therapeutic use , Male , Mitochondria/metabolism , Rabbits , Stroke/drug therapy , Tissue Plasminogen Activator/therapeutic use
11.
Acta Neurochir Suppl ; 121: 7-12, 2016.
Article in English | MEDLINE | ID: mdl-26463915

ABSTRACT

Development of drugs and devices for the treatment of stroke is not exempt from current translational research standards, which include Stroke Treatment Academic Industry Roundtable (STAIR) criteria and RIGOR guidelines. Near-infrared laser therapy (NILT) was developed to treat stroke in an era when STAIR criteria were not adhered to, thus NILT was not optimized in multiple species, nor was it optimized for efficacy across barriers in translational animal models before proceeding to expensive and extensive clinical trials. Moreover, the majority of rodent studies did not adhere to RIGOR guidelines. This ultimately led to failure in the NeuroThera Effectiveness and Safety Trial-3. Because NILT remains a promising therapeutic approach to treat stroke, we designed a systematic study to determine laser light penetration profiles across the skull of four different species with increasing skull thickness: mouse, rat, rabbit, and human.Our study demonstrates that NILT differentially penetrates the skulls. There is especially extensive attenuation of light energy penetration across the human calvaria, compared with animal skulls, which suggests that the power density setting used in stroke clinical trials may not have optimally stimulated neuroprotection and repair pathways. The results of our study suggest that NILT cannot be sufficiently optimized in "small" animals and directly translated to humans because of significant variances of skull thickness and penetration characteristics across species. NILT neuroprotection should be further studied using a research design that endeavors to incorporate human skull characteristics (thickness) into the development plan to increase the probability of success in stroke victims.


Subject(s)
Low-Level Light Therapy/methods , Skull/anatomy & histology , Stroke/therapy , Adenosine Triphosphate , Animals , Disease Models, Animal , Electron Transport Complex IV , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mitochondria , Organ Size , Rabbits , Rats , Rats, Sprague-Dawley , Tomography, X-Ray Computed
12.
J Neurol Neurophysiol ; 6(5)2015 Oct.
Article in English | MEDLINE | ID: mdl-26693049

ABSTRACT

Until recently there was little understanding of the exact pathophysiology and treatment choices for stroke patients with Pseudobulbar affect (PBA). PBA is typically characterized by outbursts or uncontrollable laughing or crying and in the majority of patients, the outbursts being involuntary and incompatible with the patients' emotional state. PBA is a behavioral syndrome reported to be displayed in 28-52% of stroke patients with first or multiple strokes, and incidence may be higher in patients who have had prior stroke events, and higher in females. There is typically involvement of glutaminergic, serotoninergic and dopaminergic neuronal circuits of the corticolimbic-subcorticothalamic-pontocerebellar network. PBA is now understood to be a disinhibition syndrome in which specific pathways involving serotonin and glutamate are disrupted or modulated causing reduced cortical inhibition of a cerebellar/brainstem-situated "emotional" laughing or crying focal center. Stroke-induced disruption of one or more neuronal pathway circuits may "disinhibit" voluntary laughing and crying making the process involuntary. With a "new" treatment currently being marketed to treat PBA patients, this article will delve into the neurological and physiological basis for PBA in stroke, and review progress with the diagnosis and treatment of PBA.

13.
Transl Stroke Res ; 6(5): 345-54, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26314402

ABSTRACT

Today, there is an enormous amount of excitement in the field of stroke victim care due to the recent success of MR. CLEAN, SWIFT PRIME, ESCAPE, EXTEND-IA, and REVASCAT endovascular trials. Successful intravenous (IV) recombinant tissue plasminogen activator (rt-PA) clinical trials [i.e., National Institute of Neurological Disorders and Stroke (NINDS) rt-PA trial, Third European Cooperative Acute Stroke Study (ECASSIII), and Third International Stroke study (IST-3)] also need to be emphasized. In the recent endovascular and thrombolytic trials, there is statistically significant improvement using both the National Institutes of Health Stroke Scale (NIHSS) and the modified Rankin Score (mRS) scale, but neither approach promotes complete recovery in patients enrolled within any particular NIHSS or mRS score tier. Absolute improvement (mRS 0-2 at 90 days) with endovascular therapy is 13.5-31 %, whereas thrombolytics alone also significantly improve patient functional independence, but to a lesser degree (NINDS rt-PA trial 13 %). This article has 3 main goals: (1) first to emphasize the utility and cost-effectiveness of rt-PA to treat stroke; (2) second to review the recent endovascular trials with respect to efficacy, safety, and cost-effectiveness as a stroke treatment; and (3) to further consider and evaluate strategies to develop novel neuroprotective drugs. A thesis will be put forth so that future stroke trials and therapy development can optimally promote recovery so that stroke victims can return to "normal" life.


Subject(s)
Brain Ischemia/drug therapy , Brain Ischemia/surgery , Embolectomy , Stroke/drug therapy , Stroke/surgery , Thrombolytic Therapy , Tissue Plasminogen Activator/therapeutic use , Aged , Aged, 80 and over , Clinical Trials as Topic , Female , Humans , Male , Middle Aged , Neuroprotection , Recombinant Proteins/therapeutic use , Reperfusion , Treatment Outcome
14.
BMC Neurol ; 15: 127, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26228060

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is associated with vascular risk factors; brain ischemia facilitates the pathogenesis of AD. Recent studies have suggested that the reduction of AD risk with statin was achieved by decreased amyloidogenic amyloid precursor protein. METHODS: We used mitochondrial transgenic neuronal cell (cybrid) models to investigate changes in the levels of intracellular hypoxia inducible factor 1α (HIF-1α) and ß-site amyloid precursor protein cleaving enzyme (BACE) in the presence of simvastatin. Sporadic AD (SAD) and age-matched control (CTL) cybrids were exposed to 2% O2 and incubated with 1 µM or 10 µM simvastatin. RESULTS: There was no significant difference between cell survival by 1 or 10 µM simvastatin in both SAD and CTL cybrids. In the presence of 1 µM simvastatin, intracellular levels of HIF-1α and BACE decreased by 40-70% in SAD, but not CTL cybrids. However, 10 µM simvastatin increased HIF-1α and BACE expression in both cybrid models. CONCLUSION: Our results suggest demonstrate differential dose-dependent effects of simvastatin on HIF-1α and BACE in cultured Alzheimer's disease cybrid cells.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Gene Expression Regulation/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Simvastatin/therapeutic use , Alzheimer Disease/pathology , Cell Line, Tumor , Cell Survival , Cells, Cultured , DNA, Mitochondrial/metabolism , Dose-Response Relationship, Drug , Humans , Hypoxia , Immunoassay , Mitochondria/pathology , Neurons/metabolism , Risk Factors
15.
PLoS One ; 10(6): e0127580, 2015.
Article in English | MEDLINE | ID: mdl-26039354

ABSTRACT

BACKGROUND AND PURPOSE: Transcranial near-infrared laser therapy (TLT) is a promising and novel method to promote neuroprotection and clinical improvement in both acute and chronic neurodegenerative diseases such as acute ischemic stroke (AIS), traumatic brain injury (TBI), and Alzheimer's disease (AD) patients based upon efficacy in translational animal models. However, there is limited information in the peer-reviewed literature pertaining to transcranial near-infrared laser transmission (NILT) profiles in various species. Thus, in the present study we systematically evaluated NILT characteristics through the skull of 4 different species: mouse, rat, rabbit and human. RESULTS: Using dehydrated skulls from 3 animal species, using a wavelength of 800nm and a surface power density of 700 mW/cm2, NILT decreased from 40.10% (mouse) to 21.24% (rat) to 11.36% (rabbit) as skull thickness measured at bregma increased from 0.44 mm in mouse to 0.83 mm in rat and then 2.11 mm in rabbit. NILT also significantly increased (p<0.05) when animal skulls were hydrated (i.e. compared to dehydrated); but there was no measurable change in thickness due to hydration. In human calvaria, where mean thickness ranged from 7.19 mm at bregma to 5.91 mm in the parietal skull, only 4.18% and 4.24% of applied near-infrared light was transmitted through the skull. There was a slight (9.2-13.4%), but insignificant effect of hydration state on NILT transmission of human skulls, but there was a significant positive correlation between NILT and thickness at bregma and parietal skull, in both hydrated and dehydrated states. CONCLUSION: This is the first systematic study to demonstrate differential NILT through the skulls of 4 different species; with an inverse relationship between NILT and skull thickness. With animal skulls, transmission profiles are dependent upon the hydration state of the skull, with significantly greater penetration through hydrated skulls compared to dehydrated skulls. Using human skulls, we demonstrate a significant correlation between thickness and penetration, but there was no correlation with skull density. The results suggest that TLT should be optimized in animals using novel approaches incorporating human skull characteristics, because of significant variance of NILT profiles directly related to skull thickness.


Subject(s)
Brain Diseases/surgery , Infrared Rays , Laser Therapy , Skull , Animals , Female , Humans , Male , Mice , Rabbits , Rats
16.
Transl Stroke Res ; 6(5): 399-406, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25990352

ABSTRACT

Spinal cord ischemia (SCI) is a devastating complication of aortic operations. Neuromonitoring using motor evoked potentials (MEPs) is a sensitive modality to detect SCI in humans. We describe a leporine SCI model using MEPs to test pharmaceutical therapeutics and other neuroprotective adjuncts. In 80 rabbits, methods to obtain MEPs in normotensive and ischemic rabbits were developed. The effects of isoflurane, propofol, apnea, and hypotension on lower extremity MEPs were studied. Lower extremity MEPs disappear upon SCI induction in 78 of 78 (100 %) rabbits. Prior to SCI induction and during apneic episodes, lower extremity MEPs were lost in all (100 %) and upper extremity MEPs in one (25 %). Isoflurane was used in four experiments, with loss of lower extremity MEPs in all four (100 %) and loss of upper extremity MEPs in zero. With propofol upper extremity, MEPs were obtainable in 80 of 80 rabbits (100 %) and lower extremity MEPs in 78 of 80 rabbits (97.5 %) prior to SCI induction. The presence of these lower extremity MEPs prior to SCI induction was not correlated with systolic or diastolic blood pressure. Disappearance of MEPs occurred in all 45 rabbits with postoperative lower extremity impairment. MEPs in the leporine model correlate closely with paraplegia. MEPs are influenced by inhaled anesthetics and apnea but not by hypotension alone. Propofol anesthesia provides reliable MEPs. This study provides the basis for a reproducible model of SCI to be used for novel therapeutic drug development.


Subject(s)
Disease Models, Animal , Evoked Potentials, Motor , Spinal Cord Ischemia/physiopathology , Anesthetics, Inhalation/administration & dosage , Anesthetics, Intravenous/administration & dosage , Animals , Apnea/chemically induced , Apnea/physiopathology , Evoked Potentials, Motor/drug effects , Hypotension/chemically induced , Hypotension/physiopathology , Isoflurane/administration & dosage , Lower Extremity/innervation , Lower Extremity/physiopathology , Propofol/administration & dosage , Rabbits , Upper Extremity/innervation , Upper Extremity/physiopathology
18.
Exp Neurol ; 266: 143-6, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25708986

ABSTRACT

OBJECTIVE: Tissue plasminogen activator (tPA) is administered to acute ischemic stroke victims in a vehicle formulation containing high concentrations of L-arginine (3.5g/100mg vial), a well-known nitric oxide synthase (NOS) substrate and precursor to nitric oxide (NO), as well as an enhancer of cerebral blood flow. METHODS: We studied the effects of tPA vehicle compared to tPA (3.3mg/kg) formulated in the same vehicle containing L-arginine, normal saline or normal saline containing L-arginine, on behavioral function following small clot embolic strokes in rabbits using clinical rating scores and quantal analysis curves as the primary end point. Treatments were administered intravenously (1ml/kg; 20% bolus/80% infused over 30min) starting 1h following the injection of small-sized blood clots into the brain vasculature and terminal behavior was measured 2days following embolization. Behavioral rating scores were used to calculate the effective stroke dose (P50 in mg) that produces neurological deficits in 50% of the rabbits. RESULTS: In this study, tPA significantly (p=0.001) improved behavior compared to all other treatments including tPA vehicle, saline and saline-L-arginine, increasing the P50 by 141% over tPA vehicle. Saline-L-arginine was not significantly different from either saline or tPA vehicle (p>0.05). CONCLUSION: This study demonstrates that the L-arginine component of the tPA vehicle does not contribute to the reproducible clinical improvement observed following tPA administration in rabbits. Moreover, the administration of L-arginine was not an effective method to promote behavioral recovery following embolic strokes in the stringent rabbit small clot stroke model, nor did L-arginine exacerbate behavioral deficits or intracerebral hemorrhage in embolized rabbits.


Subject(s)
Arginine/therapeutic use , Embolization, Therapeutic/methods , Fibrinolytic Agents/therapeutic use , Intracranial Embolism/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Embolization, Therapeutic/psychology , Intracranial Embolism/complications , Intracranial Embolism/psychology , Intracranial Hemorrhages/chemically induced , Intracranial Hemorrhages/epidemiology , Intracranial Hemorrhages/psychology , Male , Rabbits , Stroke/drug therapy , Stroke/etiology , Treatment Outcome
19.
J Neurol Neurophysiol ; 5(4)2014 Aug.
Article in English | MEDLINE | ID: mdl-25364620

ABSTRACT

Current state-of-the-art acute ischemic stroke clinical trials are designed to study neuroprotectants when administered following thrombolysis; tissue plasminogen activator (tPA) is administered to patients within 3-4.5 hours of an ischemic event. Thus, in order to develop a novel neuroprotectant and move it forward to a clinical trial, it is important to assess the effects of the drug on tPA's proteolytic activity in vitro, prior to extensive in vivo analysis. In this study, we determined if CNB-001 [4-((1E)-2-(5-(4-hydroxy-3-methoxystyryl-)-1-phenyl-1H-pyrazoyl-3-yl)vinyl)-2-methoxy-phenol)], would affect, either enhance or inhibit tPA activity in vitro. In this tPA-inhibitor (plasminogen activator inhibitor-1; PAI-1 and 2,7-Bis-(4-Amidinobenzylidene)-Cycloheptan-1-One Dihydrochloride; tPA stop) controlled study, we used a chromogenic substrate (CH3SO2-D-hexahydrotyrosine-Gly-Arg-p-nitroanilide•AcOH) to study drug interactions in vitro, spectrophotometrically measuring protease released p-Nitroaniline from the substrate. We found that PAI-1 (0.25 µM) and tPA stop (5 µM) significantly (p<0.0001) inhibited substrate release, by 98.6% and 83.4%, respectively, thus inhibiting tPA activity in vitro. In comparison, CNB-001 (0.7-7 µM) reduced tPA activity by 28-32%, with an extrapolated IC50 value of 65.2-704 µM. Thus, although high concentrations of CNB-001 does affects tPA activity in vitro, the study supports the use of CNB-001 in combination with tPA to treat stroke, However, CNB-001 should be administered following thrombolysis to promote neuroprotection and repair.

20.
J Neurol Neurophysiol ; 5(5)2014 Sep 29.
Article in English | MEDLINE | ID: mdl-25364621

ABSTRACT

Protein-Tyrosine Phosphatase1B (PTP1B) is a negative regulator of the insulin signaling pathway and is a potential therapeutic target for treatment of type 2 diabetes, cardiovascular disease, metabolic syndrome and cancer. It has been postulated that CNB-001 [4-((1E)-2-(5-(4-hydroxy-3-methoxystyryl-)-1-phenyl-1H-pyrazoyl-3-yl) vinyl)-2-methoxy-phenol)] may regulate PTP1B activity suggested by a computer-based active site docking recognition model. This possibility was studied using a human recombinant PTP1B assay, and a phospho-peptide fragment of the insulin receptor ß subunit domain (IR5). The positive control, suramin, inhibited PTP1B with an IC50 (half minimal (50%) inhibitory concentration) value of 16.34 µM; CNB-001 did not affect enzyme activity across the range of 1nM-0.1mM. This study suggests that PTP1B inhibition is not involved in the beneficial effects of CNB-001 in obese type 2 diabetic mice.

SELECTION OF CITATIONS
SEARCH DETAIL
...