Depression in Parkinson’s disease is related to a genetic polymorphism of the cannabinoid receptor gene (CNR1).

Abstract

“Depression is a common symptom in Parkinson’s disease (PD) and it is present in up to 40% of the patients. The cause of depression in PD is thought to be related to disturbance of monoamine neurotransmission. The endogenous cannabinoid system mediates different brain processes that play a role in the control of behaviour and emotions. Cannabinoid function may be altered in neuropsychiatry diseases, directly or through interactions with monoamine, GABA and glutamate systems. For this reason, we have investigated whether there is a genetic risk factor for depression in PD linked to the polymorphisms of CB1 receptor gene. Depression was more frequent in patients with PD than in controls with osteoarthritis. The presence of depression did not correlate with the stage of the disease but it was more frequent in patients with pure akinetic syndrome than in those with tremoric or mixed type PD. The CB1 receptor gene polymorphism (AAT)n is considered to modify the transcription of the gene and, therefore, it may have functional relevance. We analysed the length of the polymorphic triplet (AAT)n of the gene that encodes CB1 (CNR1) receptor in 89 subjects (48 PD patients and 41 controls). In patients with PD, the presence of two long alleles, with more than 16 repeated AAT trinucleotides in the CNR1 gene, was associated with a reduced prevalence of depression (Fisher’s exact test: P=0.003). This association did not reach significant differences in the control group, but the number of control individuals with depression was too small to allow for statistical analysis. Since the alleles with long expansions may have functional impact in cannabinoid neurotransmission, our data suggest that the pharmacological manipulation of cannabinoid neurotransmission could open a new therapeutic approach for the treatment of depression in PD and possibly in other conditions.”

http://www.ncbi.nlm.nih.gov/pubmed/15668727

Variation in the human cannabinoid receptor CNR1 gene modulates gaze duration for happy faces.

  “From an early age, humans look longer at preferred stimuli and also typically look longer at facial expressions of emotion, particularly happy faces. Atypical gaze patterns towards social stimuli are common in autism spectrum conditions (ASC). However, it is unknown whether gaze fixation patterns have any genetic basis. In this study, we tested whether variations in the cannabinoid receptor 1 (CNR1) gene are associated with gaze duration towards happy faces. This gene was selected because CNR1 is a key component of the endocannabinoid system, which is involved in processing reward, and in our previous functional magnetic resonance imaging (fMRI) study, we found that variations in CNR1 modulate the striatal response to happy (but not disgust) faces. The striatum is involved in guiding gaze to rewarding aspects of a visual scene. We aimed to validate and extend this result in another sample using a different technique (gaze tracking).

One of the key molecular systems involved in the functioning of the striatal circuit is the endocannabinoid system. It is a neuropeptidergic circuit involved in reward processing and works in tandem with the mesolimbic dopaminergic system. Expressed selectively in the brain, the cannabinoid receptor 1 (CNR1) is the best-studied molecule of this system.

This finding suggests a role for CNR1 in social reward processing and could have significance for clinical conditions such ASC, which are marked by a deficit in social reward processing as well as atypical responses to facial expressions of emotion.”

http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3155489/

Variations in the human cannabinoid receptor (CNR1) gene modulate striatal responses to happy faces.

Abstract

“Happy facial expressions are innate social rewards and evoke a response in the striatum, a region known for its role in reward processing in rats, primates and humans. The cannabinoid receptor 1 (CNR1) is the best-characterized molecule of the endocannabinoid system, involved in processing rewards. We hypothesized that genetic variation in human CNR1 gene would predict differences in the striatal response to happy faces. In a 3T functional magnetic resonance imaging (fMRI) scanning study on 19 Caucasian volunteers, we report that four single nucleotide polymorphisms (SNPs) in the CNR1 locus modulate differential striatal response to happy but not to disgust faces. This suggests a role for the variations of the CNR1 gene in underlying social reward responsivity. Future studies should aim to replicate this finding with a balanced design in a larger sample, but these preliminary results suggest neural responsivity to emotional and socially rewarding stimuli varies as a function of CNR1 genotype. This has implications for medical conditions involving hypo-responsivity to emotional and social stimuli, such as autism.”

http://www.ncbi.nlm.nih.gov/pubmed/16623851

Reduced cannabinoid CB(1) receptor binding in alcohol dependence measured with positron emission tomography.

Abstract

“Brain cannabinoid CB(1) receptors contribute to alcohol-related behaviors in experimental animals, but their potential role in humans with alcohol dependence is poorly understood. We measured CB(1) receptors in alcohol dependent patients in early and protracted abstinence, and in comparison with control subjects without alcohol use disorders, using positron emission tomography and [(18)F]FMPEP-d(2), a radioligand for CB(1) receptors. We scanned 18 male in-patients with alcohol dependence twice, within 3-7 days of admission from ongoing drinking, and after 2-4 weeks of supervised abstinence. Imaging data were compared with those from 19 age-matched healthy male control subjects. Data were also analyzed for potential influence of a common functional variation (rs2023239) in the CB(1) receptor gene (CNR1) that may moderate CB(1) receptor density. On the first scan, CB(1) receptor binding was 20-30% lower in patients with alcohol dependence than in control subjects in all brain regions and was negatively correlated with years of alcohol abuse. After 2-4 weeks of abstinence, CB(1) receptor binding remained similarly reduced in these patients. Irrespective of the diagnostic status, C allele carriers at rs2023239 had higher CB(1) receptor binding compared with non-carriers. Alcohol dependence is associated with a widespread reduction of cannabinoid CB(1) receptor binding in the human brain and this reduction persists at least 2-4 weeks into abstinence. The correlation of reduced binding with years of alcohol abuse suggests an involvement of CB(1) receptors in alcohol dependence in humans.”

http://www.ncbi.nlm.nih.gov/pubmed/22776901

Cannabinoid receptor 1 gene is associated with alcohol dependence.

Abstract

“BACKGROUND:

Alcohol dependence (AD) vulnerability is determined by a complex array of genetic factors. Given the potential role of endocannabinoid system in AD, polymorphisms within cannabinoid receptor 1 gene (CNR1) have been potentially associated with susceptibility to this disease. We thus aimed to examine the relationship between 3 allelic variants of CNR1 (rs6454674, rs1049353, and rs806368) and AD.

METHODS:

Genotyping of the aforementioned polymorphisms was carried out by PCR in 298 male alcoholics (187 of them with AD) and 155 healthy controls. Single-marker, haplotype, and interaction analysis were performed to analyze the influence of CNR1 gene on AD susceptibility.

RESULTS:

We found an association between CNR1 gene and AD after haplotype analysis. Alcoholic patients with TGT haplotype (corresponding to rs6454674-rs1049353-rs806368 polymorphisms in this order) were less prone to have AD (p = 0.017). Besides, alcoholics with a G/T substitution of the first marker (GGT haplotype) or a C/T substitution of the third marker (TGC haplotype) were more likely to develop AD (p = 0.006 and 0.004, respectively) and an interaction was found between the G allele of rs6454674 single nucleotide polymorphism (SNP) and the C allele of rs806368 SNP (p = 0.009).

CONCLUSIONS:

Our findings support previously reported associations of CNR1 with dependence to alcohol and other substances and emphasizes the relevance of endocannabinoid system in AD.”

http://www.ncbi.nlm.nih.gov/pubmed/22085192

Modulation Of The Endo-Cannabinoid System: Therapeutic Potential Against Cocaine Dependence

 “Dependence on cocaine is still a main unresolved medical and social concern, and in spite of research efforts, no pharmacological therapy against cocaine dependence is yet available. Recent studies have shown that the endocannabinoid system participates in specific stages and aspects of drug dependence in general, and some of this evidence suggests an involvement of the cannabinoid system in cocaine effects. For example, cocaine administration has been shown to alter brain endocannabinoid levels, and the endocannabinoid system has been involved in long-term modifications of brain processes that might play a role in neuro/behavioral effects of psychostimulant drugs like cocaine. Human studies show that marijuana dependence is frequently associated with cocaine dependence, and that the cannabinoid receptor CNR1 gene polymorphism might be related to cocaine addiction. This article will review the main papers in the field showing how a modulation of different components of the cannabinoid system might interact with some of the neurobiological/behavioral effects of cocaine related to its reinforcing effects, evaluated in preclinical models or in clinical settings. The goal of this review will be to provide insights into the complex picture of cocaine abuse and addiction, and to extrapolate from such endocannabinoid-cocaine interactions useful information to test the therapeutic potential of cannabinoid ligands and endocannabinoid-level enhancers against cocaine dependence for future preclinical/clinical trials.”

“Summary and concluding remarks

Most of the scientific articles reviewed in the present manuscript have described studies of cannabinoid CB1 receptor agonists and antagonists tested against cocaine effects in preclinical models thought to be predictive of cocaine abuse. These studies have provided interesting results, especially for the ability of cannabinoid antagonists, and Rimonabant in particular, to significantly counteract some of the reinforcing actions of cocaine (104, 143). Collectively, the studies suggest that a cannabinoid tone, impaired by cannabinoid antagonist administration, is indeed involved in many of the reinforcing effects of cocaine which are believed to be responsible for cocaine abuse and addiction. On the other hand, there are no studies available showing interactions of drugs acting as cannabinoid levels modulators/enhancers on cocaine-induced behaviors. These drugs affecting directly the endogenous cannabinoid tone could interfere with cocaine effects in these preclinical procedures and could substantially increase our knowledge about the cannabinoid-neurobiology related to cocaine dependence.”

“Suggestions about possible genetic predisposition/vulnerability to cocaine dependence from human studies due to variants of the cannabinoid receptor CNR1 gene have given more strength to the link between endocannabinoids and cocaine. Due to the widespread distribution of cannabinoid receptors in the brain, and their abundance in brain areas playing pivotal roles in drug abuse and addiction, the different expression and regulation of cannabinoid receptors induced by genetic differences might be an important factor in the predisposition or vulnerability to drug dependence. For this reason, the potential to directly interact with endocannabinoid tone in selected brain areas, an effect that can be obtained with endocannabinoid uptake inhibitors or metabolism blockers (as shown also in genetically modified mice, 170), as compared to widespread actions of cannabinoid receptors agonists/antagonists, should be one of the next challenges in the research for medications able to counteract the abuse- and dependence-related behavioral/neurobiological effects of cocaine.”

http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2134985/

Marijuana withdrawal and craving: influence of the cannabinoid receptor 1 (CNR1) and fatty acid amide hydrolase (FAAH) genes

 “While cannabis withdrawal may not include some of the serious medical problems observed with alcohol and opioid withdrawal, it is likely that the symptoms associated with cannabis withdrawal (e.g. negative affect, appetite and sleep disturbance) contribute to the development and intractability of cannabis dependence. In this sense, cannabis withdrawal may be analogous to other, better-understood withdrawal syndromes (e.g. tobacco withdrawal, alcohol withdrawal) that have been the target of intervention efforts. Furthermore, cannabis withdrawal has been described increasingly in terms of the physiological sequelae that coincide with this syndrome, including alterations in dopamine neurotransmission, as well as alterations in other systems.”.

“These findings may have both etiological and treatment implications. For example, individuals with the CNR1 T/C genotype may be more likely to develop dependence and/or more likely to have trouble establishing abstinence or reducing marijuana use. However, longitudinal studies will be needed to clarify whether this genetic variable actually influences the trajectory of marijuana use/dependence. In addition, treatment studies that incorporate this information are needed to determine whether these (or other) genetic variants may influence treatment outcomes and determine whether alternative treatments may be indicated for these individuals.”

“In conclusion, the cannabis dependence endophenotypes, craving and withdrawal, are important factors in the etiology and treatment of cannabis dependence and, given growing recognition of the underlying physiological sequalae that coincide with long-term cannabis use, these phenotypes are likely to lend themselves to the identification of underlying genetic factors that have direct implications for treatment approaches.”

http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2873690/

The genetic basis of the endocannabinoid system and drug addiction in humans.

Abstract

“The cannabinoid receptor (CNR1) and the fatty acid amide hydrolase (FAAH) genes are located on chromosomes 6 and 1 in the 6q15 and 1p33 cytogenetic bands, respectively. CNR1 encodes a seven-transmembrane domain protein of 472 amino acids, whereas FAAH encodes one transmembrane domain of 579 amino acids. Several mutations found in these genes lead to altered mRNA stability and transcription rate or a reduction of the activity of the encoded protein. Increasing evidence shows that these functional mutations are related to dependence upon cocaine, alcohol, marijuana, heroin, nicotine and other drugs. One of the most compelling associations is with the C385A single nucleotide polymorphism (SNP), which is found in the FAAH gene. For all of the genetic polymorphisms reviewed here, it is difficult to form overall conclusions due to the high diversity of population samples being studied, ethnicity, the use of volunteers, heterogeneity of the recruitment criteria and the drug addiction phenotype studied. Care should be taken when generalizing the results from different studies. However, many works have repeatedly associated polymorphisms in the CNR1 and FAAH genes with drug-related behaviours; this suggests that these genes should be examined in further genetic studies focusing on drug addiction and other psychiatric disorders.”

http://www.ncbi.nlm.nih.gov/pubmed/21937688

Cannabinoid receptors in brain: pharmacogenetics, neuropharmacology, neurotoxicology, and potential therapeutic applications.

“Much progress has been achieved in cannabinoid research. A major breakthrough in marijuana-cannabinoid research has been the discovery of a previously unknown but elaborate endogenous endocannabinoid system (ECS), complete with endocannabinoids and enzymes for their biosynthesis and degradation with genes encoding two distinct cannabinoid (CB1 and CB2) receptors (CBRs) that are activated by endocannabinoids, cannabinoids, and marijuana use.

Physical and genetic localization of the CBR genes CNR1 and CNR2 have been mapped to chromosome 6 and 1, respectively. A number of variations in CBR genes have been associated with human disorders including osteoporosis, attention deficit hyperactivity disorder (ADHD), posttraumatic stress disorder (PTSD), drug dependency, obesity, and depression. Other family of lipid receptors including vanilloid (VR1) and lysophosphatidic acid (LPA) receptors appear to be related to the CBRs at the phylogenetic level. The ubiquitous abundance and differential distribution of the ECS in the human body and brain along with the coupling to many signal transduction pathways may explain the effects in most biological system and the myriad behavioral effects associated with smoking marijuana. The neuropharmacological and neuroprotective features of phytocannabinoids and endocannabinoid associated neurogenesis have revealed roles for the use of cannabinoids in neurodegenerative pathologies with less neurotoxicity. The remarkable progress in understanding the biological actions of marijuana and cannabinoids have provided much richer results than previously appreciated cannabinoid genomics and raised a number of critical issues on the molecular mechanisms of cannabinoid induced behavioral and biochemical alterations. These advances will allow specific therapeutic targeting of the different components of the ECS in health and disease.

 This review focuses on these recent advances in cannabinoid genomics and the surprising new fundamental roles that the ECS plays in the retrograde signaling associated with cannabinoid inhibition of neurotransmitter release to the genetic basis of the effects of marijuana use and pharmacotherpeutic applications and limitations. Much evidence is provided for the complex CNR1 and CNR2 gene structures and their associated regulatory elements. Thus, understanding the ECS in the human body and brain will contribute to elucidating this natural regulatory mechanism in health and disease.”

http://www.ncbi.nlm.nih.gov/pubmed/19897083

‘Cannabis’ receptor discovery may help understanding of obesity and pain

“Aberdeen scientists believe that the findings—published in the —might help our understanding of these conditions and also be a step towards the development of personalised therapies to help treat them.

The team from the University’s Kosterlitz Centre for Therapeutics studied around the gene CNR1. This gene produces what are known as cannabinoid receptors, which are found in the brain, and which activate parts of the brain involved in memory, mood, appetite and pain.

activate these areas of the brain when they are triggered by chemicals produced naturally in our bodies called .

Chemicals found in the drug cannabis mimic the action of these endocannabinoids and there is growing evidence that cannabis has pain relieving and anti-inflammatory properties which can help treat diseases such as and arthritis. 

In order to understand more about these side effects and the which determine how people respond, the scientists studied genetic differences around the CNR1 gene.

Dr Alasdair MacKenzie, who helped lead the team, said: “We chose to look at one specific genetic difference in CNR1 because we know it is linked to and addiction. What we found was a mutation that caused a change in the genetic switch for the gene itself—a switch that is very ancient and has remained relatively unchanged in overthree hundred million years of evolution, since before the time of the dinosaurs.”

http://phys.org/news/2012-08-cannabis-receptor-discovery-obesity-pain.html