Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
J Pharmacol Exp Ther ; 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39134424

ABSTRACT

Neuropathic pain is a form of chronic pain that develops because of damage to the nervous system. Treatment of neuropathic pain is often incompletely effective, and most available therapeutics have only moderate efficacy and present side effects that limit their use. Opioids are commonly prescribed for the management of neuropathic pain despite equivocal results in clinical studies and significant abuse potential. Thus, neuropathic pain represents an area of critical unmet medical and novel classes of therapeutics with improved efficacy and safety profiles are urgently needed. The cannabidiol (CBD) structural analogue and novel antagonist of GPR55, KLS-13019, was screened in rat models of neuropathic pain. Tactile sensitivity associated with chemotherapy exposure was induced in rats with once daily 1mg/kg paclitaxel injections for 4 days or 5 mg/kg oxaliplatin every third day for one week. Rats were then administered KLS-13019 or comparator drugs on day 7 in an acute dosing paradigm or days 7-10 in a chronic dosing paradigm and mechanical or cold allodynia was assessed. Allodynia was reversed in a dose-dependent manner in the rats treated with KLS-13019, with the highest dose reverting the response to pre-paclitaxel injection baseline levels with both I.P. and P.O. administration after acute dosing. In the chronic dosing paradigm, 4 consecutive doses of KLS-13019 completely reversed allodynia for the duration of the phenotype in control animals. Additionally, co-administration of KLS -13019 with paclitaxel prevented the allodynic phenotype from developing. Together, these data suggest that KLS-13019 represents a potential new drug for the treatment of neuropathic pain. Significance Statement Chemotherapy-induced neuropathic pain (CIPN) is a common, debilitating side effect of cancer treatment with no known cure. GPR55 antagonist KLS-13019 represents a novel class of drug for this condition that is a potent, durable inhibitor of allodynia associated with CIPN in rats in both prevention and reversal dosing paradigms. This novel therapeutic approach addresses a critical area of unmet medical need.

2.
J Mol Neurosci ; 74(2): 41, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38602576

ABSTRACT

KLS-13019 was reported previously to reverse paclitaxel-induced mechanical allodynia in a mouse model of chemotherapy-induced peripheral neuropathy (CIPN). Recent studies demonstrated that paclitaxel-induced increases in inflammatory markers (GPR55, NLRP3, and IL-1ß) of dorsal root ganglion (DRG) cultures were shown to be reversed by KLS-13019 treatment. The mechanism of action for KLS-13019-mediated reversal of paclitaxel-induced neuroinflammation now has been explored using GPR55 siRNA. Pre-treatment of DRG cultures with GPR55 siRNA produced a 21% decrease of immunoreactive (IR) area for GPR55 in cell bodies and a 59% decrease in neuritic IR area, as determined by high-content imaging. Using a 24-h reversal treatment paradigm, paclitaxel-induced increases in the inflammatory markers were reversed back to control levels after KLS-3019 treatment. Decreases in these inflammatory markers produced by KLS-13019 were significantly attenuated by GPR55 siRNA co-treatment, with mean IR area responses being attenuated by 56% in neurites and 53% in cell bodies. These data indicate that the percentage decreases in siRNA-mediated attenuation of KLS-13019-related efficacy on the inflammatory markers were similar to the percentage knockdown observed for neuritic GPR55 IR area. Similar studies conducted with cannabidiol (CBD), the parent compound of KLS-13019, produced low efficacy (25%) reversal of all inflammatory markers that were poorly attenuated (29%) by GPR55 siRNA. CBD was shown previously to be ineffective in reversing paclitaxel-induced mechanical allodynia. The present studies indicated significant differences between the anti-inflammatory properties of KLS-13019 and CBD which may play a role in their observed differences in the reversibility of mechanical allodynia in a mouse model of CIPN.


Subject(s)
Cannabidiol , Animals , Mice , RNA, Small Interfering/genetics , Cannabidiol/pharmacology , Cannabidiol/therapeutic use , Hyperalgesia/drug therapy , Anti-Inflammatory Agents , Disease Models, Animal , Paclitaxel/toxicity , Receptors, Cannabinoid/genetics
3.
Res Sq ; 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38464007

ABSTRACT

KLS-13019 was reported previously to reverse paclitaxel-induced mechanical allodynia in a mouse model of chemotherapy-induced peripheral neuropathy (CIPN). Recent studies demonstrated that paclitaxel-induced increases in inflammatory markers (GPR55, NLRP3 and IL-1b) of dorsal root ganglion (DRG) cultures were shown to be reversed by KLS-13019 treatment. The mechanism of action for KLS-13019-mediated reversal of paclitaxel-induced neuroinflammation now has been explored using GPR55 siRNA. Pretreatment of DRG cultures with GPR55 siRNA produced a 21% decrease of immunoreactive (IR) area for GPR55 in cell bodies and a 59% decrease in neuritic IR area, as determined by high content imaging. Using a 24-hour reversal treatment paradigm, paclitaxel-induced increases in the inflammatory markers were reversed back to control levels after KLS-3019 treatment. Decreases in these inflammatory markers produced by KLS-13019 were significantly attenuated by GPR55 siRNA co-treatment, with mean IR area responses being attenuated by 56% in neurites and 53% in cell bodies. These data indicate that the percentage decreases in siRNA-mediated attenuation of KLS-13019-related efficacy on the inflammatory markers were similar to the percentage knockdown observed for neuritic GPR55 IR area. Similar studies conducted with cannabidiol (CBD), the parent compound of KLS-13019, produced low efficacy (25%) reversal of all inflammatory markers that were poorly attenuated (29%) by GPR55 siRNA. CBD was shown previously to be ineffective in reversing paclitaxel-induced mechanical allodynia. The present studies indicated significant differences between the anti-inflammatory properties of KLS-13019 and CBD which may play a role in their observed differences in the reversibility of mechanical allodynia in a mouse model of CIPN.

4.
J Mol Neurosci ; 72(9): 1859-1874, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35779192

ABSTRACT

KLS-13019, a novel devised cannabinoid-like compound, was explored for anti-inflammatory actions in dorsal root ganglion cultures relevant to chemotherapy-induced peripheral neuropathy (CIPN). Time course studies with 3 µM paclitaxel indicated > 1.9-fold increases in immunoreactive (IR) area for cell body GPR55 after 30 min as determined by high content imaging. To test for reversibility of paclitaxel-induced increases in GPR55, cultures were treated for 8 h with paclitaxel alone and then a dose response to KLS-13019 added for another 16 h. This "reversal" paradigm indicated established increases in cell body GPR55 IR areas were decreased back to control levels. Because GPR55 had previously reported inflammatory actions, IL-1ß and NLRP3 (inflammasome-3 marker) were also measured in the "reversal" paradigm. Significant increases in all inflammatory markers were produced after 8 h of paclitaxel treatment alone that were reversed to control levels with KLS-13019 treatment. Accompanying studies using alamar blue indicated that decreased cellular viability produced by paclitaxel treatment was reverted back to control levels by KLS-13019. Similar studies conducted with lysophosphatidylinositol (GPR55 agonist) in DRG or hippocampal cultures demonstrated significant increases in neuritic GPR55, NLRP3 and IL-1ß areas that were reversed to control levels with KLS-13019 treatment. Studies with a human GPR55-ß-arrestin assay in Discover X cells indicated that KLS-13019 was an antagonist without agonist activity. These studies indicated that KLS-13019 has anti-inflammatory properties mediated through GPR55 antagonist actions. Together with previous studies, KLS-13019 is a potent neuroprotective, anti-inflammatory cannabinoid with therapeutic potential for high efficacy treatment of neuropathic pain.


Subject(s)
Cannabinoids , Neuralgia , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cannabinoids/therapeutic use , Ganglia, Spinal/metabolism , Hippocampus/metabolism , Humans , NLR Family, Pyrin Domain-Containing 3 Protein , Neuralgia/drug therapy , Paclitaxel/pharmacology , Receptors, Cannabinoid/metabolism
5.
Br J Pharmacol ; 178(15): 3067-3078, 2021 08.
Article in English | MEDLINE | ID: mdl-33822373

ABSTRACT

BACKGROUND AND PURPOSE: Cannabidiol (CBD) is a non-euphorigenic component of Cannabis sativa that prevents the development of paclitaxel-induced mechanical sensitivity in a mouse model of chemotherapy-induced peripheral neuropathy (CIPN). We recently reported that the CBD structural analogue KLS-13019 shows efficacy in an in vitro model of CIPN. The present study was to characterize the behavioural effects of KLS-13019 compared to CBD and morphine in mouse models of CIPN, nociceptive pain and reinforcement. EXPERIMENTAL APPROACH: Prevention or reversal of paclitaxel-induced mechanical sensitivity were assessed following intraperitoneal or oral administration of CBD, KLS-13019 or morphine. Antinociceptive activity using acetic acid-induced stretching and hot plate assay, anti-reinforcing effects on palatable food or morphine self-administration and binding to human opioid receptors were also determined. KEY RESULTS: Like CBD, KLS-13019 prevented the development of mechanical sensitivity associated with paclitaxel administration. In contrast to CBD, KLS-13019 was also effective at reversing established mechanical sensitivity. KLS-13019 significantly attenuated acetic acid-induced stretching and produced modest effects in the hot plate assay. KLS-13019 was devoid of activity at µ-, δ- or κ-opioid receptors. Lastly, KLS-13019, but not CBD, attenuated the reinforcing effects of palatable food or morphine. CONCLUSIONS AND IMPLICATIONS: KLS-13019 like CBD, prevented the development of CIPN, while KLS-13019 uniquely attenuated established CIPN. Because KLS-13019 binds to fewer biological targets, this will help to identifying molecular mechanisms shared by these two compounds and those unique to KLS-13019. Lastly, KLS-13019 may possess the ability to attenuate reinforced behaviour, an effect not observed in the present study with CBD.


Subject(s)
Cannabidiol , Nociceptive Pain , Animals , Cannabidiol/pharmacology , Disease Models, Animal , Mice , Morphine , Reinforcement, Psychology
6.
J Mol Neurosci ; 68(4): 603-619, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31077084

ABSTRACT

Treatment with cannabidiol (CBD) or KLS-13019 (novel CBD analog), has previously been shown to prevent paclitaxel-induced mechanical allodynia in a mouse model of chemotherapy-induced peripheral neuropathy (CIPN). The mechanism of action for CBD- and KLS-13019-mediated protection now has been explored with dissociated dorsal root ganglion (DRG) cultures using small interfering RNA (siRNA) to the mitochondrial Na+ Ca2+ exchanger-1 (mNCX-1). Treatment with this siRNA produced a 50-55% decrease in the immunoreactive (IR) area for mNCX-1 in neuronal cell bodies and a 72-80% decrease in neuritic IR area as determined with high-content image analysis. After treatment with 100 nM KLS-13019 and siRNA, DRG cultures exhibited a 75 ± 5% decrease in protection from paclitaxel-induced toxicity; whereas siRNA studies with 10 µM CBD produced a 74 ± 3% decrease in protection. Treatment with mNCX-1 siRNA alone did not produce toxicity. The protective action of cannabidiol and KLS-13019 against paclitaxel-induced toxicity during a 5-h test period was significantly attenuated after a 4-day knockdown of mNCX-1 that was not attributable to toxicity. These data indicate that decreases in neuritic mNCX-1 corresponded closely with decreased protection after siRNA treatment. Pharmacological blockade of mNCX-1 with CGP-37157 produced complete inhibition of cannabinoid-mediated protection from paclitaxel in DRG cultures, supporting the observed siRNA effects on mechanism.


Subject(s)
Cannabidiol/pharmacology , Ganglia, Spinal/cytology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Sodium-Calcium Exchanger/antagonists & inhibitors , Animals , Cells, Cultured , Hyperalgesia , Neurons/metabolism , Paclitaxel/toxicity , RNA Interference , Rats , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/metabolism
7.
J Mol Neurosci ; 66(1): 121-134, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30109468

ABSTRACT

Cannabidiol (CBD) exhibits neuroprotective properties in many experimental systems. However, development of CBD as a drug has been confounded by the following: (1) low potency; (2) a large number of molecular targets; (3) marginal pharmacokinetic properties; and (4) designation as a schedule 1 controlled substance. The present work compared the properties of CBD with a novel molecule (KLS-13019) that has structural similarities to CBD. The design strategy for KLS-13019 was to increase hydrophilicity while optimizing neuroprotective potency against oxidative stress toxicity relevant to hepatic encephalopathy. The protective responses of CBD and KLS-13019 were compared in dissociated rat hippocampal cultures co-treated with toxic levels of ethanol and ammonium acetate. This comparison revealed that KLS-13019 was 31-fold more potent than CBD in preventing neuronal toxicity from the combined toxin treatment, while both compounds exhibited complete protective efficacy back to control values. In addition, treatment with KLS-13019 alone was 5-fold less toxic (TC50) than CBD. Previous studies suggested that CBD targeted the Na+-Ca2+ exchanger in mitochondria (mNCX) to regulate intracellular calcium levels, an important determinant of neuronal survival. After treatment with an inhibitor of mNCX (CGP-37157), no detectable neuroprotection from ethanol toxicity was observed for either CBD or KLS-13019. Furthermore, AM630 (CB2 antagonist) significantly attenuated CBD-mediated neuroprotection, while having no detectable effect on neuroprotection from KLS-13019. Our studies indicated KLS-13019 was more potent and less toxic than CBD. Both compounds can act through mNCX. KLS-13019 may provide an alternative to CBD as a therapeutic candidate to treat diseases associated with oxidative stress.


Subject(s)
Cannabidiol/analogs & derivatives , Cannabidiol/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Acetates/toxicity , Animals , Cells, Cultured , Ethanol/toxicity , Hippocampus/cytology , Neurons/metabolism , Rats , Sodium-Calcium Exchanger/metabolism
8.
J Mol Neurosci ; 63(1): 100-114, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28812237

ABSTRACT

Juvenile Batten disease (JBD) is an inherited disorder that is characterized by the development of blindness, seizures, and progressive motor, psychiatric, and cognitive impairment. A model of JBD expressing the predominant human mutation (Cln3 ∆ex7/8 ) has been explored. Dissociated brain cultures from Cln3 ∆ex7/8 knock-in mice were compared to wild type (WT) for effects on granules of ceroid lipofuscin (CL) and neuronal structure. Utilizing high content image analysis of CL granules identified with antibodies to mitochondrial ATP synthase subunit c or tripeptidyl peptidase-1, significant increases in the areas for both immunoreactive granules were observed in Cln3 ∆ex7/8 cultures in comparison to WT. CL granules also exhibit autofluorescence at 488 and 560 nm, and the areas of these autofluorescent spots were found to be significantly increased in Cln3 ∆ex7/8 cultures in comparison to WT. Progressive increases in CL granule area in Cln3 ∆ex7/8 cultures were observed during culture development. Because current therapies for JBD provide only symptomatic support, a therapeutic strategy has been explored based on the observations that JBD-related tissues are deficient in ß-galactosyl ceramide. Treatment of cultures for 40 h with a potent analog of ß-galactosyl ceramide (SNB-4050) produced significant decreases in CL granule area in the Cln3 ∆ex7/8 cultures; whereas identical studies on WT cultures produced no detectible changes. Significant decreases in average neurite length and neurite branch point number were also observed in the Cln3 ∆ex7/8 cultures that were attenuated by treatment with 1 nM SNB-4050. These studies indicate Cln3 ∆ex7/8 brain cultures may be useful to screen therapeutic agents for treatment of JBD.


Subject(s)
Brain/cytology , Galactosylceramides/pharmacology , Lipofuscin/metabolism , Membrane Glycoproteins/genetics , Molecular Chaperones/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Neurons/drug effects , ATP Synthetase Complexes/metabolism , Aminopeptidases/metabolism , Animals , Cells, Cultured , Cytoplasmic Granules/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Mice , Neurons/cytology , Neurons/metabolism , Serine Proteases/metabolism , Tripeptidyl-Peptidase 1
9.
ACS Med Chem Lett ; 7(4): 424-8, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27096053

ABSTRACT

Cannabidiol is the nonpsychoactive natural component of C. sativa that has been shown to be neuroprotective in multiple animal models. Our interest is to advance a therapeutic candidate for the orphan indication hepatic encephalopathy (HE). HE is a serious neurological disorder that occurs in patients with cirrhosis or liver failure. Although cannabidiol is effective in models of HE, it has limitations in terms of safety and oral bioavailability. Herein, we describe a series of side chain modified resorcinols that were designed for greater hydrophilicity and "drug likeness", while varying hydrogen bond donors, acceptors, architecture, basicity, neutrality, acidity, and polar surface area within the pendent group. Our primary screen evaluated the ability of the test agents to prevent damage to hippocampal neurons induced by ammonium acetate and ethanol at clinically relevant concentrations. Notably, KLS-13019 was 50-fold more potent and >400-fold safer than cannabidiol and exhibited an in vitro profile consistent with improved oral bioavailability.

10.
J Mol Neurosci ; 52(3): 446-58, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24277343

ABSTRACT

Severe seizure activity is associated with reoccurring cycles of excitotoxicity and oxidative stress that result in progressive neuronal damage and death. Intervention with these pathological processes is a compelling disease-modifying strategy for the treatment of seizure disorders. We have optimized a series of small molecules for neuroprotective and anticonvulsant activity as well as altered their physical properties to address potential metabolic liabilities, to improve CNS penetration, and to prolong the duration of action in vivo. Utilizing phenotypic screening of hippocampal cultures with nutrient medium depleted of antioxidants as a disease model, cell death and decreased neuronal viability produced by acute treatment with glutamate or hydrogen peroxide were prevented. Modifications to our previously reported proof of concept compounds have resulted in a lead which has full neuroprotective action at <1 nM and antiseizure activity across six animal models including the kindled rat and displays excellent pharmacokinetics including high exposure to the brain. These modifications have also eliminated the requirement for a chiral molecule, removing the possibility of racemization and making large-scale synthesis more easily accessible. These studies strengthen our earlier findings which indicate that potent, multifunctional neuroprotective anticonvulsants are feasible within a single molecular entity which also possesses favorable CNS-active drug properties in vitro and in vivo.


Subject(s)
Anticonvulsants/chemical synthesis , Neurons/drug effects , Neuroprotective Agents/chemical synthesis , Animals , Anticonvulsants/adverse effects , Anticonvulsants/pharmacokinetics , Anticonvulsants/pharmacology , Caco-2 Cells , Cells, Cultured , Enterocytes/drug effects , Humans , Mice , Mice, Inbred C57BL , Neuroprotective Agents/adverse effects , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley
11.
J Med Chem ; 56(22): 9019-30, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24205976

ABSTRACT

Broad-spectrum anticonvulsants are of considerable interest as antiepileptic drugs, especially because of their potential for treating refractory patients. Such "neurostabilizers" have also been used to treat other neurological disorders, including migraine, bipolar disorder, and neuropathic pain. We synthesized a series of sulfamide derivatives (4-9, 10a-i, 11a, 11b, 12) and evaluated their anticonvulsant activity. Thus, we identified promising sulfamide 4 (JNJ-26489112) and explored its pharmacological properties. Compound 4 exhibited excellent anticonvulsant activity in rodents against audiogenic, electrically induced, and chemically induced seizures. Mechanistically, 4 inhibited voltage-gated Na(+) channels and N-type Ca(2+) channels and was effective as a K(+) channel opener. The anticonvulsant profile of 4 suggests that it may be useful for treating multiple forms of epilepsy (generalized tonic-clonic, complex partial, absence seizures), including refractory (or pharmacoresistant) epilepsy, at dose levels that confer a good safety margin. On the basis of its pharmacology and other favorable characteristics, 4 was advanced into human clinical studies.


Subject(s)
Amides/chemistry , Amides/pharmacology , Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Dioxanes/chemistry , Dioxanes/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Absorption , Amides/pharmacokinetics , Amides/therapeutic use , Animals , Anticonvulsants/pharmacokinetics , Anticonvulsants/therapeutic use , Dioxanes/pharmacokinetics , Dioxanes/therapeutic use , Dogs , Drug Evaluation, Preclinical , Drug Resistance , Epilepsy/drug therapy , Female , Humans , Male , Mice , Rats , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use
12.
J Mol Neurosci ; 47(2): 368-79, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22535312

ABSTRACT

Severe seizure activity is associated with recurring cycles of excitotoxicity and oxidative stress that result in progressive neuronal damage and death. Intervention to halt these pathological processes is a compelling disease-modifying strategy for the treatment of seizure disorders. In the present study, a core small molecule with anticonvulsant activity has been structurally optimized for neuroprotection. Phenotypic screening of rat hippocampal cultures with nutrient medium depleted of antioxidants was utilized as a disease model. Increased cell death and decreased neuronal viability produced by acute treatment with glutamate or hydrogen peroxide were prevented by our novel molecules. The neuroprotection associated with this chemical series has marked structure activity relationships that focus on modification of the benzylic position of a 2-phenyl-2-hydroxyethyl sulfamide core structure. Complete separation between anticonvulsant activity and neuroprotective action was dependent on substitution at the benzylic carbon. Chiral selectivity was evident in that the S-enantiomer of the benzylic hydroxy group had neither neuroprotective nor anticonvulsant activity, while the R-enantiomer of the lead compound had full neuroprotective action at <40 nM and antiseizure activity in three animal models. These studies indicate that potent, multifunctional neuroprotective anticonvulsants are feasible within a single molecular entity.


Subject(s)
Anticonvulsants/pharmacology , Epilepsy/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Sulfonamides/pharmacology , Animals , Anticonvulsants/chemistry , Caco-2 Cells , Cells, Cultured , Disease Models, Animal , Epilepsy/pathology , Hippocampus/cytology , Hippocampus/drug effects , Humans , Mice , Neurons/pathology , Neuroprotective Agents/chemistry , Rats , Sulfonamides/chemistry
14.
J Med Chem ; 52(23): 7528-36, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19388676

ABSTRACT

In seeking broad-spectrum anticonvulsants to treat epilepsy and other neurological disorders, we synthesized and tested a group of sulfamide derivatives (4a-k, 5), which led to the clinical development of 4a (JNJ-26990990). This compound exhibited excellent anticonvulsant activity in rodents against audiogenic, electrically induced, and chemically induced seizures, with very weak inhibition of human carbonic anhydrase-II (IC(50) = 110 microM). The pharmacological profile for 4a supports its potential in the treatment of multiple forms of epilepsy, including pharmacoresistant variants. Mechanistically, 4a inhibited voltage-gated Na(+) channels and N-type Ca(2+) channels but was not effective as a K(+) channel opener. The pharmacokinetics and metabolic properties of 4a are discussed.


Subject(s)
Amides/chemistry , Amides/pharmacology , Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Thiophenes/chemistry , Thiophenes/pharmacology , Amides/metabolism , Amides/pharmacokinetics , Animals , Anticonvulsants/metabolism , Anticonvulsants/pharmacokinetics , Carbonic Anhydrase II/antagonists & inhibitors , Cell Line , Clinical Trials as Topic , Drug Evaluation, Preclinical , Female , Humans , Male , Mice , Rats , Sulfonamides/metabolism , Sulfonamides/pharmacokinetics , Thiophenes/metabolism , Thiophenes/pharmacokinetics
15.
J Pharmacol Exp Ther ; 328(3): 813-21, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19064719

ABSTRACT

Previous studies have shown that cathepsins control amyloid beta (Abeta) levels in chromaffin cells via a regulated secretory pathway. In the present study, this concept was extended to investigations in primary hippocampal neurons to test whether Abeta release was coregulated by cathepsins and electrical activity, proposed components of a regulated secretory pathway. Inhibition of cathepsin B (catB) activity with CA074Me or attenuation of catB expression through small interfering RNA produced decreases in Abeta release, similar to levels produced with suppression of beta-site APP-cleaving enzyme 1 (BACE1) expression. To test whether the catB-dependent release of Abeta was linked to ongoing electrical activity, neurons were treated with tetrodotoxin (TTX) and CA074Me. These comparisons demonstrated no additivity between decreases in Abeta release produced by TTX and CA074Me. In contrast, pharmacological inhibition of cathepsin L (catL) selectively elevated Abeta42 levels but not Abeta40 or total Abeta. Mechanistic studies measuring C-terminal fragments of amyloid precursor protein (APP) suggested that catL elevated alpha-secretase activity, thereby suppressing Abeta42 levels. The mechanism of catB-mediated regulation of Abeta release remains unclear but may involve elevation of beta-secretase. In summary, these studies provide evidence for a significant alternative pathway for APP processing that involves catB and activity-dependent release of Abeta in a regulated secretory pathway for primary neurons.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cathepsin B/metabolism , Cathepsins/metabolism , Cysteine Endopeptidases/metabolism , Hippocampus/physiology , Neurons/physiology , Animals , Cathepsin B/genetics , Cathepsin L , Cathepsins/genetics , Cysteine Endopeptidases/genetics , Hippocampus/enzymology , Humans , Neurons/drug effects , RNA, Small Interfering/genetics , Synapses/drug effects , Synapses/physiology , Tetrodotoxin/pharmacology
16.
Epilepsy Res ; 83(1): 66-72, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19013768

ABSTRACT

Carisbamate (RWJ-333369; (S)-2-O-carbamoyl-1-o-chlorophenyl-ethanol) is a novel investigational antiepileptic drug that exhibits a broad-spectrum of activity in a number of animal models of seizure and drug refractory epilepsy. In an effort to understand the molecular mechanism by which carisbamate produces its antiepileptic actions, we studied its effects on the function of voltage-gated, rat brain sodium and potassium channels and on the repetitive firing of action potentials in cultured rat hippocampal neurons. In whole-cell patch clamp recording, carisbamate resulted in a concentration-, voltage- and use-dependent inhibition of rat Nav1.2, with an IC(50) value of 68 microM at -67 mV. In rat hippocampal neurons, carisbamate similarly blocked voltage-gated sodium channels, with an IC(50) value of 89 microM at -67 mV, and inhibited repetitive firing of action potentials in a concentration-dependent manner (by 46% at 30 microM and 87% at 100 microM, respectively). Carisbamate had no effect on the steady-state membrane potential or voltage-gated potassium channels (K(v)) in these neurons. These inhibitory effects of carisbamate occurred at therapeutically relevant concentrations in vivo, raising the possibility that block of voltage-gated sodium channels by carisbamate contributes to its antiepileptic activity.


Subject(s)
Action Potentials/drug effects , Anticonvulsants/pharmacology , Carbamates/pharmacology , Hippocampus/physiology , Neurons/physiology , Sodium Channel Blockers/pharmacology , Animals , Cell Line , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Electrophysiology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Membrane Potentials/drug effects , Neurons/drug effects , Patch-Clamp Techniques , Phenytoin/pharmacology , Pregnancy , Rats , Rats, Sprague-Dawley
17.
J Med Chem ; 50(18): 4261-4, 2007 Sep 06.
Article in English | MEDLINE | ID: mdl-17685503

ABSTRACT

A new aspartic protease inhibitory chemotype bearing a 2-amino-3,4-dihydroquinazoline ring was identified by high-throughput screening for the inhibition of BACE-1. X-ray crystallography revealed that the exocyclic amino group participated in a hydrogen bonding array with the two catalytic aspartic acids of BACE-1 (Asp(32), Asp(228)). BACE-1 inhibitory potency was increased (0.9 microM to 11 nM K(i)) by substitution into the unoccupied S(1)' pocket.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Models, Molecular , Quinazolines/chemical synthesis , Amyloid Precursor Protein Secretases/chemistry , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/chemistry , CHO Cells , Caco-2 Cells , Cell Membrane Permeability , Cricetinae , Cricetulus , Crystallography, X-Ray , Humans , Hydrogen Bonding , Molecular Conformation , Mutation , Oligopeptides/chemistry , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/blood , Peptide Fragments/metabolism , Quinazolines/chemistry , Quinazolines/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship
18.
J Neurosci Methods ; 165(2): 175-82, 2007 Sep 30.
Article in English | MEDLINE | ID: mdl-17645948

ABSTRACT

Technical variants of mania and depression models that were based on dominant-submissive relationships (DSR) have been analyzed and compared in the present paper. In these paradigms, one animal of a pair developed the behavioral trait of dominance while the other submissiveness in a food competition test after repeated interactions in a specially designed apparatus. Data collection methods and timelines have been compared in variants of the DSR-based models. In addition, different selection criteria to assign dominant or submissive status to animals and two different scoring systems were evaluated. The importance of the selection criteria for DSR stability has been emphasized. Our data showed that (1) only animals selected with the strict criteria form clear dominant and submissive relationships that hold throughout the study period, (2) submissive animals were influenced by fluoxetine and dominant animals were influenced by sodium valproate similarly in pairs scored by human observer and by a video-tracking system. These studies indicate that the model variant using stringent selection criteria and automatic scoring was the most reliable for use in depression-related studies.


Subject(s)
Antidepressive Agents/pharmacology , Antimanic Agents/pharmacology , Behavior, Animal/drug effects , Bipolar Disorder/drug therapy , Depressive Disorder/drug therapy , Dominance-Subordination , Animals , Behavior, Animal/physiology , Bipolar Disorder/physiopathology , Bipolar Disorder/psychology , Data Interpretation, Statistical , Depressive Disorder/physiopathology , Depressive Disorder/psychology , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Feeding Behavior/physiology , Fluoxetine/pharmacology , Neuropsychological Tests/standards , Rats , Rats, Sprague-Dawley , Rats, Wistar , Selective Serotonin Reuptake Inhibitors/pharmacology , Social Behavior , Valproic Acid/pharmacology , Videotape Recording/methods
19.
Neurosci Biobehav Rev ; 31(6): 904-19, 2007.
Article in English | MEDLINE | ID: mdl-17597209

ABSTRACT

There is confusion in the literature on the measurement of the drug activity onset time (AOT) for both clinical and non-clinical studies of antidepressant and antimanic drugs. The questions asked are: How often and at which time points should drug effects be measured? At what level of a drug effect should AOT be determined? Is the placebo (control) effect important for consideration of drug AOT? This paper reviews approaches taken to answer these questions and to assess drug therapeutic AOT. The first part of the paper is devoted to a review of methods used in clinical trials with depression as an indication. The second part is focused on approaches taken in animal models of depression and how they could help in assessing drug AOT. Finally, a summary of pharmacological values on which the AOT depends is presented and a new statistical approach to data analysis method proposed. The allied experimental design for pre-clinical and clinical studies may help to characterize and differentiate AOT for available and new generation of antidepressants and antimanic drugs.


Subject(s)
Antidepressive Agents/pharmacology , Antimanic Agents/pharmacology , Behavior, Animal/drug effects , Dominance-Subordination , Reaction Time/drug effects , Animals , Bipolar Disorder/drug therapy , Depressive Disorder/drug therapy , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Humans , Time Factors
20.
Peptides ; 28(9): 1720-6, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17513014

ABSTRACT

The neuroprotective properties of vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) place these peptides in a special category of ligands that have implications for our understanding of pathological conditions as well as a potential basis for therapeutic intervention. It is remarkable that these peptides have a protective impact against such a wide variety of clinical relevant toxic substances. This protective diversity is consistent with the multiple pathways that are activated or inhibited by the action of these peptides. Although knowledge is emerging on the neuroprotective mechanisms of VIP and PACAP, it is already evident that these two peptides are not identical in their action and each peptide has multiple mechanisms that allow for neuroprotective diversity. The multiple intracellular signaling pathways and differing extracellular mediators of neuroprotection contribute to this diversity of action. In this review, examples of neuroprotective actions will be presented that serve to demonstrate the remarkable breadth of neuroprotective processes produced by VIP and PACAP.


Subject(s)
Neurons/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Vasoactive Intestinal Peptide/pharmacology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Chemokines/metabolism , Humans , Neurons/cytology , Neurons/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL