ArticlePublisher preview available

Cannabidiol Post-Treatment Alleviates Rat Epileptic-Related Behaviors and Activates Hippocampal Cell Autophagy Pathway Along with Antioxidant Defense in Chronic Phase of Pilocarpine-Induced Seizure

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Abnormal and sometimes severe behavioral and molecular symptoms are usually observed in epileptic humans and animals. To address this issue, we examined the behavioral and molecular aspects of seizure evoked by pilocarpine. Autophagy can promote both cell survival and death, but there are controversial reports about the neuroprotective or neurodegenerative effects of autophagy in seizure. Cannabidiol has anticonvulsant properties in some animal models when used as a pretreatment. In this study, we investigated alteration of seizure scores, autophagy pathway proteins, and antioxidant status in hippocampal cells during the chronic phase of pilocarpine-induced epilepsy after treatment with cannabidiol. Cannabidiol (100 ng, intracerebroventricular injection) delayed the chronic phase of epilepsy. Single administration of cannabidiol during the chronic phase of seizure significantly diminished seizure scores such as mouth clonus, head nodding, monolateral and bilateral forelimb clonus and increased the activity of catalase enzyme and reduced glutathione content. Such a protective effect in the behavioral scores of epileptic rats was also observed after repeated administrations of cannabidiol at the onset of the silent phase. Moreover, the amount of Atg7, conjugation of Atg5/12, Atg12, and LC3II/LC3I ratio increased significantly in epileptic rats treated with repeated injections of cannabidiol. In short, our results suggest that post-treatment of Cannabidiol could enhance the induction of autophagy pathway and antioxidant defense in the chronic phase of epilepsy, which could be considered as the protective mechanisms of cannabidiol in a temporal lobe epilepsy model.
This content is subject to copyright. Terms and conditions apply.
Cannabidiol Post-Treatment Alleviates Rat Epileptic-Related
Behaviors and Activates Hippocampal Cell Autophagy Pathway
Along with Antioxidant Defense in Chronic Phase
of Pilocarpine-Induced Seizure
Mahshid Hosseinzadeh
1
&Sara Nikseresht
1
&Fariba Khodagholi
1,2
&Nima Naderi
1,3
&
Nader Maghsoudi
1,2
Received: 11 November 2015 /Accepted: 15 December 2015 /Published online: 6 January 2016
#Springer Science+Business Media New York 2016
Abstract Abnormal and sometimes severe behavioral and
molecular symptoms are usually observed in epileptic humans
and animals. To address this issue, we examined the behav-
ioral and molecular aspects of seizure evoked by pilocarpine.
Autophagy can promote both cell survival and death, but there
are controversial reports about the neuroprotective or neuro-
degenerative effects of autophagy in seizure. Cannabidiol has
anticonvulsant properties in some animal models when used
as a pretreatment. In this study, we investigated alteration of
seizure scores, autophagy pathway proteins, and antioxidant
status in hippocampal cells during the chronic phase of
pilocarpine-induced epilepsy after treatment with cannabidiol.
Cannabidiol (100 ng, intracerebroventricular injection) de-
layed the chronic phase of epilepsy. Single administration of
cannabidiol during the chronic phase of seizure significantly
diminished seizure scores such as mouth clonus, head nod-
ding, monolateral and bilateral forelimb clonus and increased
the activity of catalase enzyme and reduced glutathione con-
tent. Such a protective effect in the behavioral scores of epi-
leptic rats was also observed after repeated administrations of
cannabidiol at the onset of the silent phase. Moreover, the
amount of Atg7, conjugation of Atg5/12, Atg12, and LC3II/
LC3I ratio increased significantly in epileptic rats treated with
repeated injections of cannabidiol. Inshort, our results suggest
that post-treatment of Cannabidiol could enhance the induc-
tion of autophagy pathway and antioxidant defense in the
chronic phase of epilepsy, which could be considered as the
protective mechanisms of cannabidiol in a temporal lobe ep-
ilepsy model.
Keywords Pilocarpine-induced seizure .Cannabidiol .
Autophagy .Antioxidant status
Introduction
One of the most common forms of partial epilepsy in humans is
temporal lobe epilepsy (TLE) (Engel, 2001). Characterization
of this model shows three phases: (a) a period of 24 h is known
as the acute phase that extended to limbic area and causes status
epilepticus (SE), (b) the second phase is a silent period where
electroencephalogram and behavior are both normal and vary
from 4 to 44 days, and (c) a third period known as chronic
phase is characterized by spontaneous recurrent seizures
(SRSs) (Cavalheiro, 1995,Aridaetal.1999). Localization of
seizure foci in the limbic system, particularly in the hippocam-
pus, is the main feature of TLE (Bartolomei et al. 2005).
Autophagy, one of the most important pathways that main-
tain cell homeostasis, can regulate important biological func-
tions such as cell survival, cell death, cell metabolism, devel-
opment, neuroprotection, and sometimes neurodegeneration
(Hochfeld et al. 2013). In this process, cytoplasmic compo-
nents are delivered to lysosomes to form autophagosomes. In
this process, a group of autophagy-related proteins (Atgs)
have vital roles (Levine and Klionsky, 2004,Meijer,2003).
Among these proteins, Atg5 initiates the process (Maiuri et al.
2007). The participation of two ubiquitin-like conjugation
*Nader Maghsoudi
nmaghsoudi@sbmu.ac.ir
1
Neuroscience Research Center, Shahid Beheshti University of
Medical Sciences, Tehran, Iran
2
NeuroBiology Research Center, Shahid Beheshti University of
Medical Sciences, Tehran, Iran
3
Department of Pharmacology and Toxicology, School of Pharmacy,
Shahid Beheshti University of Medical Sciences, Tehran, Iran
J Mol Neurosci (2016) 58:432440
DOI 10.1007/s12031-015-0703-6
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
... In alignment with these findings, Giacoppo and colleagues [49] reported that CBD influences the PI3K/Akt/mTOR pathway and enhances neuroprotection by inhibiting JNK and p38 MAP kinases (Figure 2). Furthermore, Hosseinzadeh et al. [50] found that repeated intracerebroventricular injections of 0.100 ng CBD in epileptic rats resulted in the activation of various autophagy markers, including the conjugation of Atg5/12, Atg7, Atg12, and the expression of LC3-II/LC3-I, particularly in hippocampal cells, thereby confirming the protective effects associated with the autophagy pathway. ...
Chapter
Full-text available
The dysregulation of autophagy plays a significant role in the onset of various pathologies, with emerging evidence indicating its potential involvement in chronic pain conditions. The cellular and molecular mechanisms underlying this pathology have been thoroughly investigated, with the endocannabinoid system (ECS) identified as a crucial factor in the progression of chronic neuropathic pain. This chapter highlights several plant-derived cannabinoids, including cannabidiol, cannabinol, Δ-9-tetrahydrocannabinol, which exhibit unique modulatory effects on the ECS. We discuss the roles of cannabinoid receptors, cannabinoid enzymes, and PPARγ as modulators in decreased levels of inflammatory cytokines, as well as a reduction in microglial activation, and inhibit the assembly of NLRP3 inflammasome complex, which potentially provides substantial neuroprotective effects in neuropathic pain. Since the discovery of the endocannabinoid system and the implications of mitochondrial dysfunction and autophagy impairment in neuropathic pain, there has been an increasing interest in the therapeutic potential of this system.
... However, only 100 µM CBD significantly reduced burst amplitude. These findings are in line with the previous literature stating CBD's potential biphasic effect depending on the feature of interest [7,59]. ...
Article
Full-text available
Cannabinoids have shown potential in drug-resistant epilepsy treatment; however, we lack knowledge on which cannabinoid(s) to use, dosing, and their pharmacological targets. This study investigated (i) the anticonvulsant effect of Cannabidiol (CBD) alone and (ii) in combination with Delta-9 Tetrahydrocannabinol (Δ⁹-THC), as well as (iii) the serotonin (5-HT)1A receptor’s role in CBD’s mechanism of action. Seizure activity, induced by 4-aminopyridine, was measured by extracellular field recordings in cortex layer 2/3 of mouse brain slices. The anticonvulsant effect of 10, 30, and 100 µM CBD alone and combined with Δ⁹-THC was evaluated. To examine CBD’s mechanism of action, slices were pre-treated with a 5-HT1A receptor antagonist before CBD’s effect was evaluated. An amount of ≥30 µM CBD alone exerted significant anticonvulsant effects while 10 µM CBD did not. However, 10 µM CBD combined with low-dose Δ⁹-THC (20:3 ratio) displayed significantly greater anticonvulsant effects than either phytocannabinoid alone. Furthermore, blocking 5-HT1A receptors before CBD application significantly abolished CBD’s effects. Thus, our results demonstrate the efficacy of low-dose CBD and Δ⁹-THC combined and that CBD exerts its effects, at least in part, through 5-HT1A receptors. These results could address drug-resistance while providing insight into CBD’s mechanism of action, laying the groundwork for further testing of cannabinoids as anticonvulsants.
... The authors suggested that this contributes to the hepatoprotective effect of CDB and Cannabis sativa. Our results showed that mainly, the catalase level was increased, even in the normoxic group, which could contribute to the beneficial effects of CBD1 and 2. Similarly, enhanced catalase activity was found in mice hippocampal tissue after CBD treatment in a pilocarpine-induced seizure rat model [40]. ...
Article
Full-text available
Cannabidiol (CBD) is a nonpsychoactive phytocannabinoid that can be found in Cannabis sativa and possesses numerous pharmacological effects. Due to these promising effects, CBD can be used in a wide variety of diseases, for instance cardiovascular diseases. However, CBD, like tetrahydrocannabinol (THC), has low bioavailability, poor water solubility, and a variable pharmacokinetic profile, which hinders its therapeutic use. Chemical derivatization of CBD offers us potential ways to overcome these issues. We prepared three new CBD derivatives substituted on the aromatic ring by Mannich-type reactions, which have not been described so far for the modification of cannabinoids, and studied the protective effect they have on cardiomyocytes exposed to oxidative stress and hypoxia/reoxygenation (H/R) compared to the parent compound. An MTT assay was performed to determine the viability of rat cardiomyocytes treated with test compounds. Trypan blue exclusion and lactate dehydrogenase (LDH) release assays were carried out to study the effect of the new compounds in cells exposed to H2O2 or hypoxia/reoxygenation (H/R). Direct antioxidant activity was evaluated by a total antioxidant capacity (TAC) assay. To study antioxidant protein levels, HO-1, SOD, catalase, and Western blot analysis were carried out. pIC50 (the negative log of the IC50) values were as follows: CBD1: 4.113, CBD2: 3.995, CBD3: 4.190, and CBD: 4.671. The newly synthesized CBD derivatives prevented cell death induced by H/R, especially CBD2. CBD has the largest direct antioxidant activity. The levels of antioxidant proteins were increased differently after pretreatment with synthetic CBD derivatives and CBD. Taken together, our newly synthesized CBD derivatives are able to decrease cytotoxicity during oxidative stress and H/R. The compounds have similar or better effects than CBD on H9c2 cells.
Article
Full-text available
Purpose This study aimed to investigate the hypothesis that cannabidiol (CBD), with known anti-inflammatory and anti-apoptotic effects, would reduce the severity of acute lung injury in pulmonary contusion following blunt chest trauma. Methods Forty male Wistar Albino rats were randomly divided into four groups, each consisting of 10 rats: Sham, Trauma, Trauma + CBD, and CBD. The rats were treated with a single dose of 5 mg/kg CBD intraperitoneally 30 min before trauma. Then, the trauma were exposed to a weight of 200 g and a height of 1 m. After sacrifice, the lung tissues were removed for histopathological, immunohistochemical, biochemical, and genetic analyses. Results Pulmonary injury of trauma group led to increases in tumor necrosis factor α, caspase-3, caspase-9, Bcl-2-associated X protein expressions, total oxidant status, oxidative stress index levels, and decreases in B-cell lymphoma expression and total antioxidant levels. Additionally, inflammatory cell infiltration, damage-related emphysema, pronounced hyperemia, and increased septal tissue thickness were observed histopathologically. CBD treatment ameliorated all these findings. Conclusion CBD reduces lung damage in lung contusions caused by blunt chest trauma through its anti-inflammatory and antiapoptotic effects. More detailed studies investigating other important intracellular pathways are needed.
Chapter
In the last two decades, the endocannabinoid system has emerged as a crucial modulator of motivation and emotional processing. Due to its widespread neuroanatomical distribution and characteristic retrograde signaling nature, cannabinoid type I receptors and their endogenous ligands finely orchestrate somatic and axon terminal activity of dopamine neurons. Owing to these unique features, this signaling system is a promising pharmacological target to ameliorate dopamine-mediated drug-seeking behaviors while circumventing the adverse side effects of, for instance, dopaminergic antagonists. Despite considerable preclinical efforts, an agreement on the efficacy of endocannabinoid-targeting compounds for treating drug substance use disorders in humans has not been reached. In the following chapter, we will summarize preclinical and clinical evidence addressing the therapeutic potential of cannabinoids and endocannabinoid-targeting compounds in substance use disorders. To bridge the gap between animal and clinical research, we capitalize on studies evaluating the impact of endocannabinoid-targeting compounds in relevant settings, such as the management of drug relapse. Finally, we discuss the therapeutic potential of novel cannabinoid compounds that hold promise for treating substance use disorders.
Article
Cannabis was used to treat convulsions and other disorders since ancient times. In the last few decades, preclinical animal studies and clinical investigations have established the role of cannabidiol (CBD) in treating epilepsy and seizures and support potential therapeutic benefits for cannabinoids in other neurological and psychiatric disorders. Here, we comprehensively review the role of cannabinoids in epilepsy. We briefly review the diverse physiological processes mediating the central nervous system response to cannabinoids, including D ⁹ -THC, cannabidiol, and terpenes. Next, we characterize the anti- and proconvulsive effects of cannabinoids from animal studies of acute seizures and chronic epileptogenesis. We then review the clinical literature on using cannabinoids to treat epilepsy, including anecdotal evidence and case studies as well as the more recent randomized-controlled clinical trials that led to FDA approval of CBD for some types of epilepsy. Overall, we seek to evaluate our current understanding of cannabinoids in epilepsy and focus future research on unanswered questions.
Article
Full-text available
Background Treatment-resistant epileptic seizures are associated with reduced quality of life (QoL). As polypharmacy with routine antiseizure medications has many side effects, novel add-on treatments are necessary. Recent research showed the efficacy of add-on therapy by cannabidiol (CBD) on refractory epilepsy. We attempted to extend data on the efficacy and safety profile of CBD in patients with frontal lobe treatment-resistant epilepsy. Methods A total of 27 patients were recruited into two CBD (n = 12) and placebo (n = 15) groups. The CBD group received a highly purified liposomal preparation of the drug in addition to routine antiseizure medications. The placebo group only received antiseizure medications. This experiment followed a triple-blinding protocol. Outcome measures were seizure frequency, the Chalfont seizure severity scale (CSSS), and the quality of life questionnaire score (QOLIE-31) assessed at baseline, 4 weeks, and 8 weeks. Results At 4 weeks, results indicated that a higher fraction of patients in the CBD group (66.67%) showed improvement in seizure, compared to the placebo group (20.00%). Before–after comparison revealed that CBD, unlike routine ADEs, was effective in reducing the occurrence of seizures at the study's final timepoint [mean difference 45.58, 95% CI (8.987 to 82.18), p = 0.009]. Seizure severity was not affected by study groups or time intervals (repeated-measures ANOVA p > 0.05). Post-hoc tests found that the QoLI-31 score was improved at 8 weeks compared to baseline [mean diff. −5.031, 95% CI (−9.729 to −0.3328), p = 0.032]. The difference in cases who experienced enhanced QoL was meaningful between the CBD and placebo groups at 8 weeks [RR: 2.160, 95% CI (1.148 to 4.741), p = 0.018] but not at 4 weeks (p = 0.653). A positive finding for QoL improvement was associated with a positive finding for seizure frequency reduction [r = 0.638, 95% CI (0.296 to 0.835), p = 0.001]. Interestingly, limiting the correlation analysis to cases receiving CBD indicated that QoL improvement was not linked with seizure parameters such as severity and frequency (p > 0.05). Conclusion The present study suggests the benefit of a purified and highly efficient preparation of CBD for seizure frequency reduction and improvement of QoL in refractory frontal lobe epilepsy. Further study with longer follow-ups and larger sample size is advised. Clinical trial registration https://www.irct.ir/trial/56790, identifier: IRCT20210608051515N1.
Article
Epilepsy (EP) is a long-term neurological disorder characterized by neuroinflammatory responses, neuronal apoptosis, imbalance between excitatory and inhibitory neurotransmitters, and oxidative stress in the brain. Autophagy is a process of cellular self-regulation to maintain normal physiological functions. Emerging evidence suggests that dysfunctional autophagy pathways in neurons are a potential mechanism underlying EP pathogenesis. In this review, we discuss current evidence and molecular mechanisms of autophagy dysregulation in EP and the probable function of autophagy in epileptogenesis. Moreover, we review the autophagy modulators reported for the treatment of EP models, and discuss the obstacles to, and opportunities for, the potential therapeutic applications of novel autophagy modulators as EP therapies. Teaser: Defective autophagy affects the onset and progression of epilepsy, and many anti-epileptic drugs have autophagy-modulating effects.
Article
Full-text available
Neurodegenerative disorders are generally characterized by abnormal aggregation and deposition of specific proteins. Amyloid beta (Aβ)-associated neurodegenerative disorder is characterized by an oxidative damage that, in turn, leads to some behavioral changes before the establishment of dementia such as depression and anxiety. In the current study, we investigated the effect of heat shock protein 90 inhibitor geldanamycin (GA) administration 24 h before Aβ injection. In our experiment, 7 days after Aβ injection, elevated plus maze and forced swimming test were conducted to assess anxiety and depression-like behaviors. Levels of autophagy markers and malondialdehyde (MDA) and also activity of catalase in the hippocampus of rats were evaluated. Our behavioral analyses demonstrated that GA pretreatment can significantly decrease anxiety- and depression-like behaviors in Aβ-injected rats. Also, levels of autophagy markers including Atg12, Atg7, and LC3-II increased, while MDA level decreased and the activity of catalase increased in rats pretreated with GA compared to Aβ-injected rats. Thus, we assumed that GA, at least in part, ameliorated Aβ-mediated anxiety and depression by inducing autophagy and improving antioxidant defense system.
Article
Article
A protein determination method which involves the binding of Coomassie Brilliant Blue G-250 to protein is described. The binding of the dye to protein causes a shift in the absorption maximum of the dye from 465 to 595 nm, and it is the increase in absorption at 595 nm which is monitored. This assay is very reproducible and rapid with the dye binding process virtually complete in approximately 2 min with good color stability for 1 hr. There is little or no interference from cations such as sodium or potassium nor from carbohydrates such as sucrose. A small amount of color is developed in the presence of strongly alkaline buffering agents, but the assay may be run accurately by the use of proper buffer controls. The only components found to give excessive interfering color in the assay are relatively large amounts of detergents such as sodium dodecyl sulfate, Triton X-100, and commercial glassware detergents. Interference by small amounts of detergent may be eliminated by the use of proper controls.