Open Access

Understanding the genetics and neurobiological pathways behind addiction (Review)

  • Authors:
    • Alexandra Popescu
    • Maria Marian
    • Ana Miruna Drăgoi
    • Radu-Virgil Costea
  • View Affiliations

  • Published online on: March 23, 2021     https://doi.org/10.3892/etm.2021.9976
  • Article Number: 544
  • Copyright: © Popescu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The hypothesis issued by modern medicine states that many diseases known to humans are genetically determined, influenced or not by environmental factors, which is applicable to most psychiatric disorders as well. This article focuses on two pending questions regarding addiction: Why do some individuals become addicted while others do not? along with Is it a learned behavior or is it genetically predefined? Recent data suggest that addiction is more than repeated exposure, it is the synchronicity between intrinsic factors (genotype, sex, age, preexisting addictive disorder, or other mental illness), extrinsic factors (childhood, level of education, socioeconomic status, social support, entourage, drug availability) and the nature of the addictive agent (pharmacokinetics, path of administration, psychoactive properties). The dopamine‑mesolimbic motivation‑reward‑reinforcement cycle remains the most coherent physiological theory in addiction. While the common property of addictive substances is that they are dopamine‑agonists, each class has individual mechanisms, pharmacokinetics and psychoactive potentials.

1. Introduction

Substance-related disorders are a set of behavioral, cognitive and physiological phenomena that occur after repeated use of a substance. These typically include: A strong desire to continue using a drug, difficulties in controlling its use, persistence in using it although it has negative consequences, the use of the substance taking precedence over other activities and obligations, along with high tolerance and sometimes withdrawal (1). New developments have altered the way we define addiction. In this sense the Diagnostic and Statistical Manual of Mental Disorders (5th ed.) (DSM-5) (2) has changed the related chapter from ‘Substance-Related Disorders’ to ‘Substance-Related and Addictive Disorders’ as well as it lists the following types of substance addictions: Alcohol; caffeine; tobacco; cannabis; hallucinogens; inhalants; opioids; sedatives, hypnotics and anxiolytics; and stimulants.

Formerly known as an impulse control disorder, gambling was introduced in the category of addictions within the DSM-5. This important change occurred because the pathogenic mechanisms behind gambling are more similar to substance use disorders (2). This approach was initially opposed by the International Statistical Classification of Diseases and Related Health Problems 10th Revision (ICD-10) (3), which included pathological gambling to impulse control disorders alongside compulsive sexual disorder, kleptomania, pyromania and intermittent explosive disorder. Introducing gambling disorder to ICD-11 was a point of contention; ultimately the ICD-11 reclassified pathological gambling to gambling disorder and exchanged it from habit and impulse disorders to disorders due to substance use or addictive behaviors. More so, for the first time, gaming disorder was added to disorders due to substance use or addictive behaviors within the ICD-11, a decision challenged by video game producers (4-6).

Although addiction has been classified as a disease since the late 1800s due to its debilitating nature on both the individual and on society, to this day neuroscientists and behavioral scientists have yet to reach a common conclusion on its cause. While neuroscientists seek genetic background and neurological correlates in the reward circuitry that accompany the development of addiction, behavioral scientists, on the other hand, strive to develop and attest behavioral models of addiction (7).

2. Aims and methods

This medical review has proposed the following targets: i) to highlight that addiction has a genetic determinant, which under the influence of both internal factors and external factors, is activated after exposure to addictive agents, giving rise to addiction; ii) to summarize the neurobiological pathways associated with the chronic relapsing seen in addiction-neurotransmitters and reward circuits; and iii) to call attention to the importance of the nature of addictive agents (pharmacokinetics, psychoactive properties) in developing addiction.

We conducted a systematic review by gathering information from PubMed database on the subject of addiction in reference to the aforementioned questions. The selected articles have been published within the last 15 years. The main key words used were: ‘Addiction genetics’, ‘reward system’, ‘dopamine’, ‘learned behavior’ and ‘drugs’.

3. Genetics and epigenetics

Addiction is a multifactorial process and it is difficult to understand why some individuals are more susceptible to developing addictive behavior than others. An individual's background, moral codes and social status determine whether someone may become an addict, but also a person's genetics is one of the most important factors in the development of addiction as far as modern medicine dictates. Heritability is responsible for 40-60% of the population's variability in developing an addiction (8).

There has been proof of both genetic factors that influence the susceptibility of developing an addiction in general, and other genes and sets of genes more specific for one substance or type of addiction (9).

Until genome-wide association studies (GWASs), genetic variant associations were not substantially established. GWASs compare the DNA of individuals that have different phenotypes for a specific trait or medical condition with a control group formed by similar individuals without the disease. Thus, GWASs identify single nucleotide polymorphisms (SNPs), as well as other DNA variants, that are associated with a disease. Unfortunately, SNPs explain only a part of the variance in substance addiction and further research is required. The first GWAS conducted on the subject of addiction was regarding nicotine dependence (10-12).

The relationship between genetic influences and environmental factors took center stage in terms of new findings. It has been pointed out that these two factors can modulate each other (13). For example, one study concluded that genetic influences were decreased in adolescent smoking twins when the parental monitoring increased (11). More so, childhood adversity, stressful life events and lower levels of education seem to have an effect over alcohol-metabolizing, dopaminergic and serotonin transporter genes (9).

Epigenetics studies the heritable changes in phenotype that do not occur after DNA sequence alterations. DNA methylation and modifications of histones are the most studied epigenetic alterations. There are also epigenetic enzymes that mediate DNA demethylation and have important roles in learning, memory, neurodevelopment, but also in some psychiatric and neurologic disorders (14). There are studies regarding epigenetic changes in the molecular processes that result in addiction to psychostimulants (15,16). Repeated stressful life events are capable of causing epigenetic changes. Given that addicts are individuals with stressful lives, this may explain why these individuals are more vulnerable to neuroplastic changes induced by drugs, changes that constitute the substrate of addiction (17).

Research conducted on mice has emphasized the influence of epigenetic alterations on resilient phenotypes. Thus, in contrast to susceptible mice, resilient ones did not have alterations in the expression of the G9a histone methyltransferase enzyme in their nucleus accumbens when experiencing chronic stress (18).

Furthermore, evidence suggests that maternal conduct can have an epigenetically mediated impact on the offspring's hypothalamic-pituitary-adrenal response to stress (19). Such evidence has been found also regarding the link of paternal stress and increased DNA methylation in the offspring's hippocampus (20).

By identifying the coping styles of resilient individuals and mentoring people at risk for substance use disorder, it is possible to transmit resilient behaviors across generations with the help of epigenetics, in order to gain an important benefit for the population and the health care system.

4. Neurotransmitters

Most known neurotransmitters seem to be involved in addiction in different ways and at different moments.

Dopamine (3,4-dihydroxytyramine)

Mounting evidence places the dopaminergic-mesolimbic system as the leading system in the development of addiction, due to its role in encoding motivation and reward (21).

Apart from the motivation-reward-reinforcement cycle, the dopaminergic system plays numerous other roles such as executive functioning and motor planning, sleep and regulation of food intake, neuroendocrine secretion, arousal and sexual gratification. An imbalance in any function may lead to significant disorders. Several neuropsychiatric disorders (Parkinson's disease, Huntington disease, Alzheimer's disease, schizophrenia, bipolar disorder, attention-deficit disorders, Tourette's syndrome) have been associated with a variation in dopamine (22).

Studies have described five types of dopamine receptors: D1, D2, D3, D5, D4 (ordered by density) of the G-protein coupled receptor type. By studying the functioning of these five receptors two subclasses have been characterized: D-1 like receptors (D1, D5) and D-2 like receptors (D2, D3, D4). D1-like receptors activate adenylate cyclase, converting ATP to cAMP, in order to disinhibit protein kinase A, which phosphorylates cAMP regulatory element binding protein (CREB). Thus D1-like receptors have a vital role in the regulation of the reward system, learning, and memory. Furthermore, D1 receptors have been linked to various neuropsychiatric disorders, by activating the phospholipase C and inducing intracellular calcium release. In contrast, binding dopamine to D-2 like receptors inhibits adenylate cyclase which decreases the production of cAMP (23).

From a neurophysiological point of view, addiction can be translated as a hypo-dopaminergic dysfunctional state within the reward circuitry, and therefore is characterized by a decreased in dopamine D2 receptors. Moreover, a greater risk of addiction is associated with the polymorphism of Taq1A (rs1800497), responsible for the number of D2 receptors, the A1 allele having a lower density of D2 receptors (21,24).

Rebalancing dopamine is a difficult objective to reach in order to treat addiction. Using antipsychotics has shown some beneficial results for isolated alcoholism, while in the subpopulation of stimulant users it may aggravate the condition (25).

Serotonin (5-hydroxytryptamine, 5-HT)

Available data demonstrate the complex roles of serotonin such as regulating neuroplasticity, cognition and memory, behavior and mood, social behavior and sexual desire, impulse control, as well as appetite, sleep, circadian rhythmicity and neuroendocrine functions (26). Analyzing this assemblage of functions, a simple conclusion can be drawn: Comorbid mood and addictive disorders are expected due to dysregulation of serotonin. When discussing serotonin, 5-HT2CR (serotonin receptor) cannot be overlooked, as it is recognized as an important nominator in depression, suicide, sexual dysfunction, addiction and obesity (27,28).

5-HT has been linked to addiction by several studies (26,29). Additionally, the serotonergic response is believed to differ from the initial exposure to chronic use, from development of dependence to withdrawal, from abstinence to relapse. For example, during substance intake, 5-HT levels increase which is correlated with a boost in mood while in withdrawal syndrome there is hypoactivity of the serotonin system which may contribute to dysphoria (26-29).

Recent research has examined the role of 5-HT in impulse control, as high levels of impulsivity may be considered a risk factor for the vulnerability to addiction and relapse (29).

Whether to treat the underlying depression, ameliorate withdrawal symptoms, or reduce craving, the efficiency of using antidepressants in addiction disorders is still being debated by physicians. Certain antidepressants, such as selective serotonin reuptake inhibitors (SSRIs), can be used for mood elevation during detox, while bupropion is used to reduce nicotine cravings and ease withdrawal symptoms (29,30).

Endogenous opioids (endorphin, encephalin)

Endogenous opioids are natural pain killers, much like exogenous opioids. Additionally, they control motor activity, intestinal tract motility and peristalsis, the hypothalamic neuroendocrine axis and limbic system regulation of emotional behavioral, modulating euphoric responses and opposing stress (31-33). These effects glorify opioids in substance users. The euphoria together with their highly addictive properties are a recipe for disaster. Endorphins impact addiction via two routes, either by stimulating the mesolimbic dopamine system also independently, by an increase in endorphins in the extracellular space in the nucleus accumbens (34).

Naltrexone, a subtype-nonspecific opioid receptor antagonist, is currently used to treat opioid addiction and alcoholism. Some studies propose naltrexone as a pan-addiction treatment, yet more research is needed (34,35).

Acetylcholine (ACH)

The cholinergic system plays a supporting role in encoding motivation alongside dopamine, but it is also involved in sensory and motor processing, sleep, nociception, mood, stress response, attention, arousal and memory (36).

In regards to addiction, ACH is involved by processing reward, acquisitioning conditional associations and conditioned learning, satiation and aversion, drug procurement through arousal and attention. Learning and memory are essential to repeated behaviors (37,38).

Considering its implication, cholinergic medications may represent a potential treatment for addiction but further research is needed. However, AChE inhibitors (donepezil, rivastigmine) can be considered for cognitive impairment associated with long-term substance use (37).

γ-aminobutyric acid) (GABA)

GABA is the prime inhibitory neurotransmitter; in normal conditions it regulates fear and anxiety. A decrease in cerebral GABA has been linked to schizophrenia, depression, anxiety, addiction and sleep disorders (39).

A decreased activation in the GABAA receptor induces tolerance when exposed to chronic doses of alcohol, while the GABAB receptor is involved in adjunct treatments to aid in the initiation of abstinence, maintenance of abstinence, and prevention of cue-related relapse in some addictions (40).

Baclofen, a GABA derivative and GABAB agonist, has been proposed as treatment for alcohol disorders, yet additional research is warranted (41).

Glutamate

Glutamate is the most substantial excitatory neurotransmitter and is known for the following roles: Synaptic plasticity, cognition, learning and memory, and emotions. Glutamatergic dysfunction has been associated with a series of neuropsychiatric disorders including depression, anxiety, bipolar disorder, schizophrenia and addiction disorder.

Chronic substance use is responsible for plastic changes in glutamatergic response in striatum and midbrain dopamine neurons, intensifying the brain's reactivity to drugs (42).

Ketamine and esketamine are antagonist of the N-methyl-D-aspartate (NMDA) glutamate receptor. Preliminary data suggest that ketamine can prolong abstinence from alcohol and heroin as well as reduce the craving for cocaine, but ketamine itself may lead to addiction if used ill-advisedly exceeding the therapeutical dose (43).

5. Reward circuit

The reward system is a collection of brain structures and neural pathways that are responsible for: Associative learning (primarily classical conditioning and operator strengthening); motivation and desire; emotions with a positive value, especially pleasure (44).

The ventral tegmental area (VTA) is the preface of the reward circuit; it is composed mainly of dopamine neurons (60-65%) alongside GABA neurons 30-35% and some glutamatergic neurons (2-3%). To simplify things, dopamine is released by the VTA and is directed toward dopamine receptors (D-1 and D-2 like) established in the nucleus accumbens (NAc) via the mesolimbic pathway, prefrontal cortex (PFC) via the mesocortical pathways as well as other brain regions such as the amygdala and hippocampus. In this cascade, each cerebral structure plays a complex role in inducing addiction. The nucleus accumbens is a conglomeration of dopamine-sensitive neurons that strongly contributes to the motivation-reward-reinforcement cycle through positive reinforcement and pleasure. The prefrontal cortex plays a complex role in planning action to obtain reward-related substances. The hippocampus is involved in the formation of new memories, in order to remember and seek out pleasurable stimuli. These memories are consolidated with the help of the amygdala, also associated with emotion (44-46).

GABA neurons are vital for processing reward, with similar connections, but opposing effects, GABA activation inhibits dopamine release from the VTA, consequently decreasing dopamine throughout the entire circuit, having a strong role in drug aversion (45).

Dopamine does not act alone; it is influenced by the cholinergic system on three levels: a) VTA: Both receptors (nAChR and mAChR) stimulate the secretion of dopamine; b) NAc: The stimulus that translates into reward is transmitted over an extensive network of neurons between the cortical and subcortical area via cholinergic interneurons; c) PFC: The cholinergic system regulates cognition and conditioned learning setting base for addiction (36,37).

The best example of how the reward circuit functions is based on a legendary study performed on rats, which quickly learn to press a lever to obtain the desired dose of electrical stimulation, pressing the lever thousands of times/hour until exhausted (47). Later on, Goeders and their team allowed rats to self-administer opioids directly into the medial prefrontal increasing dopamine in the nucleus accumbens. In this perspective, animals, like humans, engage in behaviors that increase the release of dopamine (48). The following information will be treated as a new subtitle below (See Point 8 Psychopathology).

6. Addictive agents

Addictive substances act as dopamine-agonists; it is the one common property that ties these agents together otherwise each class has individual mechanisms, pharmacokinetics and psychoactive potentials, which will be treated separately below.

Alcohol

Alcohol addiction is a global burden; therefore, it has been immensely studied throughout the decades. Repeated administration of ethanol is the cause of neurological alteration within the circuits that control motivational processes. This leads to affecting how arousal, reward, and stress are encoded in the brain, creating a vicious cycle. Additionally, chronic use of alcohol induces a change in the aforementioned neurotransmitters leading to sensitization and tolerance.

One neurotransmitter that undergoes change is dopamine. Once acquainted with alcohol, the nucleus accumbens will increase dopamine activity in the anticipation of the substance reinforcing repeated consumption. In reverse, withdrawal produces a decrease in dopamine function, which may contribute to withdrawal symptoms and alcohol relapse (34).

Secondarily, alcoholism affects the expression of GABAA genes resulting in substance tolerance (27,32). Other genes identified that are involved in the metabolism of alcohol are ADH1B and ALDH2, including GABRA2, CHRM2, KCNJ6 and AUTS2. These genes increase the risk of alcoholism or related traits (49).

Thirdly, a difference in the behavioral responses to stress has been described; alcohol use produces a dysregulation in the hypothalamic-pituitary-adrenal axis (34).

Moreover, alcoholism causes neuroimmune gene induction which alters the limbic system and frontocerebellar neuronal nodes contributing to persistent drinking (50).

Multiple imaging techniques have been utilized to observe the dynamic metabolic changes and imbalance of neurotransmitter accelerating neurodegenerative alteration that occur after chronic exposure to ethanol. Examining the data presented by Sullivan and Pfefferbaum, it was found that both frontal lobe and connective circuitry suffered modifications due to the chronic intake of alcohol (51).

Future research must attempt to fully understand the genetic impact behind alcohol dependency in order to discover genetically tailored treatment for alcoholism.

Tobacco

Nicotine is at the root of tobacco addiction, one of the most abused substances in the world due to its legal status and easy access. Few genetic components have been described in nicotine addiction, especially in adolescent smokers (52). Nicotine binds to nicotinic cholinergic receptors (nAChRs), mediating the complex actions of nicotine in tobacco users. CYP2A6 is responsible for metabolizing nicotine and variability in the metabolic rate contributes to the susceptibility of tobacco dependence, withdrawal symptoms and the risk of lung cancer (53,54). Much like other substances, repeated nicotine exposure alters sensitivity to dopamine within the reward network and circuits involved in learning, stress, and self-control. One particularity of nicotine is that levels peak after 10 sec of inhalation reaching a quicker and faster ‘endorphin high’ in comparisons to other drugs which translates to frequent cravings and relapses (55).

Electrochemical signaling modifications in the anterior frontal lobe as well as atrophy and neurodegenerative disease have been linked to chronic smoking. In regards to smoking tobacco, a close look to the vascular changes is called for, because the associated decreased cerebral blood perfusion can be a determinant factor in cognitive dysfunction. Moreover, in addition to nicotine, cigarettes contain a multitude of potentially cytotoxic compounds hence singling out a sole cause of atrophy would be challenging (55).

Hallucinogens

Serotonergic hallucinogens or psychedelics are highly psychoactive substances that are associated with perceptual disorders such as hallucinations, complex cognitive symptoms and mood disturbances. The most common representatives from this class are LSD (d-lysergic acid diethylamide), psilocybin (4-phosphoryloxy-N, N-dimethyltryptamine), peyote (mescaline), DMT (dimethyltryptamine), and ayahuasca. The method by which psychedelics function is through binding to serotonin 5-HT2A receptors which is associated with increased cortical glutamate levels. Correspondingly, neuroimaging has demonstrated an increase in prefrontal cortical metabolism. Recreational use of psychedelics is more frequent than addiction although with repeated exposure tolerance sets in rapidly due to downregulation of 5-HT2A receptors (56).

Early studies have sought to use hallucinogens as agonists or partial agonists of 5-HT to alleviate severe forms of depression (57).

Inhalants

Inhalants define a class of substances based on the route of administration (inhalatory or breathing in volatile chemical vapors). Individually, the representatives of this class have different pharmacokinetics. These substances include solvents, aerosol, sprays, gases and nitrites.

Intoxication from acute volatile substances can vary from alcohol-like effects with stimulation or loss of inhibition to intense euphoria and hallucinations, depending on the substance and the dose. Accurate evidence on the effects of chronic inhalant administration is scarce and the results are usually influenced by polydrug use (58).

Dependence on inhalants is often explained by the nature of the substance, the easy availability, the cheap price and the faster onset of effects, yet the neurophysiological and neurochemical aspects need more documentation (59).

Cannabis

Δ9-tetrahydrocannabinol (THC) is the principal psychoactive component in cannabis; the one responsible for the addictive potential. Available data suggest that THC is a partial agonist of the CB1R (endocannabinoid receptor). Direct effects on the endocannabinoid system and indirect impact on the GABA-ergic, glutamatergic and dopaminergic systems, result in THC producing effects on emotional, executive, memory and reward processing (60).

Cannabidiol (CBD) is also synthesized by the cannabis plant and, in comparison to THC, lacks intoxicating effects, and can offset some of the acute effects of THC. Recent studies suggest it has a positive role in the treatment of epilepsy, addictions, anxiety disorders and psychosis (61,62).

Cannabis plants have become more and more selected to produce THC only in order to maintain consumption.

CB1R is a G protein-coupled receptor that exists in high concentrations in the amygdala, hippocampus, thalamus, cerebellum, basal ganglia and neocortex (especially in the limbic and frontal areas), areas associated with emotional, cognitive and reward processing. Activation of these receptors situated within the central brain reward circuits plays an important role in the pleasurable and anxiolytic effects of the drug. Researchers suggest that THC acts upon reward substrates in a seemingly way as do other abused drugs (60).

There are endogenous lipid-based retrograde neurotransmitters that form the endocannabinoid system which influences the motivation for natural rewards (social interaction, food, sexual activity) and modulates the rewarding effects of addictive drugs. Endocannabinoids bind to the CB1Rs and this leads to suppression in glutamatergic nerve terminals and suppression of inhibition in GABA-ergic nerve terminals. This signaling pathway is disrupted by THC (60).

After THC exposure, impaired salience processing has been observed, a fact that has been explained by the dysregulation of the dopaminergic and endocannabinoid systems that are involved in salience attribution (63).

Research has shown that chronic exposure to THC downregulates CB1Rs, a fact that explains the tolerance to the rewarding effects of the drug (64). Moreover, there is evidence that CB1R density is restored after one month of abstinence (except the hippocampus), so not all neurobiological changes are permanent in chronic cannabis users (65,66).

An interest in discovering a link between genetic factors and lifelong cannabis use has increased over time. There is interesting data available on this subject, but further research is needed. An association between rs1800497 Taq1A of the ANKK1 gene, the gene locus HES7/PER1 on chromosome 17 and cannabis consumption has been found (67), and parental care has also been shown to play an important role (68). More so, depression and self-harm appear to be genetically and phenotypically linked to cannabis use, but the direction of the causality requires further study (69). HTR2B is a major locus associated with cannabis-induced aggression, as it is known that cannabis increases impulsivity and decreases behavioral inhibition (70). The largest genome-wide association study pointed out the significant single nucleotide polymorphism and gene associations in 16 regions. It was shown that the CHRNA2 gene has a decreased expression in the cerebellum of cannabis-dependent people (71). Other genes associated with lifetime cannabis use are NCAM1 (implicated in alcohol use, smoking, schizophrenia, mood disorders), SCOC, CADM2 (a synaptic cell adhesion molecule from the immunoglobulin family, linked to risk-taking behavior, alcohol consumption, processing speed) and KCNT2 (72,73).

Opioids

Even though effective treatment is available for opioid intoxication, relapse is frequent in opioid addicts, mainly due to the opioid receptors' tolerance after repeated use of opioids. After tolerance is developed and the euphoria fades, addicts start feeling symptoms of withdrawal, such as abdominal cramps, diarrhea, sweating, agitation, bone pain, myalgia, and rhinorrhea (74). These withdrawal symptoms develop quickly and can be alleviated by correct treatment.

Opioid receptors are located in the brain, skin, spinal cord, gastrointestinal tract and after stimulation cause euphoria, analgesia, sedation and respiratory depression.

There are three discovered subtypes of opioid receptors: Mu, kappa and delta, with different effects (the common effect is analgesia) and disposition. Mu receptor stimulation produces euphoria, respiratory depression and physical dependence; kappa receptors trigger sedation and dysphoria, while delta receptors stimulate anxiolysis. Mu receptors are crucial for the activation of the reward system (75). An interesting explanation for why adolescents are more predisposed to addictive behaviors in comparison with adults lies in the fact that in this category of patients mu opioid receptors have increased positive reinforcement and less withdrawal symptoms (76).

Studies have concluded that kappa opioid receptors have anti-reward effects in opposition to mu receptors. They are involved in the relapse of addicts, because during the addiction process these receptors are stimulated, leading to dysphoria in withdrawal and abstinence phases, and finally to relapse (77,78).

In order to stave off withdrawal symptoms without consuming more heroin, patients can receive opioid agonist therapy with methadone, buprenorphine or naloxone. Methadone is a full mu opioid receptor, which also has some agonist effects on kappa and delta receptors. Its half-life is longer, causes fewer withdrawal symptoms and helps opioid addicts have a more stable life. Buprenorphine is a partial mu agonist, partial kappa agonist or functional antagonist, delta agonist and is safer than methadone. Naloxone is an opioid receptor antagonist used for acute opioid intoxications (79).

Researchers have attempted to discover genes linked to opioid addiction and a genome-wide association study discovered SNPs from multiple loci-KCNG2*rs62103177 are connected to opioid dependence (80).

Genes associated with heroin addiction have been classified into two systems: The dopaminergic one and the mu opioid receptor one. In the dopaminergic gene system, the following SNPs are listed: rs1800497, rs1079597, rs4680, rs747302, rs936462, rs1800498, rs1800955, while in the mu opioid receptor gene system, rs7997012, rs1799971 and rs540825 are included (81).

Sedatives, hypnotics, and anxiolytics

Benzodiazepines (BZDs) are the most representative class for this category of drugs. BZDs have been increasingly prescribed by doctors in the last few years, thus BZD abuse and dependence have become a serious medical issue (82).

The calming or sedating effect of BZDs occurs after binding to a specific site on the GABA type A receptors (ligand-gated ion channels), the excitatory neurons are inhibited, the VTA glutaminergic drive is reduced, as well as the nucleus accumbens' dopamine release. The GABA-ergic neurotransmission is modulated by chronic exposure to BZDs, which leads to tolerance, dependence and withdrawal. KB220 is a pro-dopamine regulator, that can be used to produce dopamine homeostasis and combat benzodiazepine use disorder, but future research is needed in this field (83,84).

There have been attempts to discover genetic factors that play a role in BZD addiction. Researchers have investigated the genetic polymorphisms of MAOA (the gene metabolizing catecholamine) and GABA A subunit alpha 2, because of their potential link with anxiety and addiction. The results have been unsatisfactory, as none of the investigated polymorphisms did determine addiction. However, studies revealed some genetic predispositions to personality features. For example, genotype 3/3 MAOA is associated with lack of anxiety and higher extraversion, while genotype 4/4 MAOA was found in individuals with higher levels of introversion and anxiety (85). Given these data, the genetic implication in BZD dependence remains a subject for further exploration.

Stimulants Cocaine

Cocaine hydrochloride is used especially intranasally, but it can also be administered subcutaneous and intravenous or by smoking crack cocaine. The path of administration is related to the severity of the dependence; therefore higher levels of addiction have been described in injection users, followed by smokers, and lower levels with intranasal users (86). Its effects include euphoria, increased sexual appetite, enhancement of intellectual and physical activity, higher self-esteem and easier social networking, which makes cocaine an attractive substance. It has been linked with high economic status and male to female ratio (87). Like in the case of other substances that cause dependence, environmental risk factors are important in cocaine addiction. Some of those factors include childhood abuse, peer drug use, drug availability, household drug use and poor social activities (86). Interestingly, parental monitoring is not important for cocaine use (88).

Regarding genetics, research indicates that there is little specific genetic variance for cocaine addiction. One genome-wide association study on cocaine addiction identified an SNP which maps to an intron of the FAM53B gene. This gene is believed to be linked with axonal extension during development and cell proliferation (89).

Some research suggests a hereditary factor in cocaine addiction. Several studies regarding the effects of paternal cocaine use on offspring behavior conclude that individuals from the next generation were more likely to consume drugs of abuse (90).

The medical community is awaiting the discovery of an effective treatment for cocaine addiction, such as a cocaine vaccine (91), cocaine hydrolase, or even deep brain stimulation of the nucleus accumbens (92).

Methamphetamine (METH)

Although METH is a highly addictive drug, its addiction mechanisms are less studied.

Available data suggest that METH exposure activates neuroinflammatory and neuroplastic processes in the brain, which may lead to parkinsonism (secondary to DA neuron damage), cognitive deficits, depression and promote addiction (93).

Researchers have found important decreases in DA levels, density of dopamine transporters, tyrosine hydroxylase levels in both the striatum and the cortex of METH addicts (94).

More so, imaging studies have shown a reactive astrogliosis inside the brain of METH abusers. The microgliosis has been shown to precede changes in striatal dopamine neurons, thus suggesting that microglial activation is implicated in the development of neurodegeneration (94). Microgliosis was found to persist two years after the beginning of abstinence and has been linked to long-term neurological damage of METH (95).

Studies have aimed to ascertain whether METH addiction can be caused by epigenetically induced alterations in gene expression known to play a role in cognitive functions and synaptic plasticity. It has been found that this drug upregulates several genes that are involved in cell-to-cell signaling and in cAMP response element-binding protein (CREB), such as GIRK2, HCRTR1, GABBR2, and KCNJ6. Activation of GIRK and GABBR2 mediate DA neuronal excitability (96).

7. Behavioral addictions

Gambling addiction and gaming disorder are the single two non-substance addiction introduced in the ICD-11. Other behavior addictions yet to be officially recognized by the medical community include food, sex, pornography, shopping, and exercise.

The overexpression of ΔFosB in the nucleus accumbens seems to be present in both behavioral addiction (food, sex, exercise) as well as substance addiction (alcohol, cannabinoids, cocaine, nicotine, and amphetamines) (97). An even closer look shows that gambling disorder and gaming disorder have a similar genetic background known as DRD2 Taq1A1, specifically the Ankk1 mutation (98).

Behavioral addictions follow the same reward patterns as substance use, with the same associated phenomena of craving, tolerance, relapse and withdrawal (described as anxiety, irritability, and emotional instability) (99).

Gambling disorder (GD)

Based on the ICD-11 criteria for gambling disorder (4), gambling disorder is characterized by a pattern of persistent or recurrent gambling behavior, which may be online (i.e., over the internet) or offline, manifested by: i) impaired control over gambling (e.g., onset, frequency, intensity, duration, termination, context); ii) increasing priority given to gambling to the extent that gambling takes precedence over other life interests and daily activities; and iii) continuation or escalation of gambling despite the occurrence of negative consequences.

The behavior pattern is of sufficient severity to result in significant impairment in personal, family, social, educational, occupational or other important areas of functioning. The pattern of gambling behavior may be continuous or episodic and recurrent. The gambling behavior and other features are normally evident over a period of at least 12 months in order for a diagnosis to be assigned, although the required duration may be shortened if all diagnostic requirements are met and symptoms are severe.

The severity of gambling can be predicted with two main personality traits found in gamblers: Harm avoidance and self-directedness. Additionally, the inability to regulate negative emotions has also been associated with a risk of non-strategic gambling (100). Approximately 96% of gambling addicts have overlapped criteria with at least one other psychiatric diagnosis, and 49% have been treated for another mental illness (101). The study of neuroimaging in gamblers shows structural and functional modifications in the reward circuits (102).

Gambling disorder presents 50-60% heritability rates (103). Furthermore, genetic findings were described in gambling disorder such as the associations of the C/C genotype of the serotonin receptor 2A T102C (rs 6313) polymorphism and the PG phenotype (103).

A nuclear medicine tomographic imaging technique (SPECT) found an interesting parallel between psychostimulant drugs such as amphetamines and a motorbike-riding computer game regarding the ventral striatum's dopamine release (104). It is known that video game addiction in adolescence is a disguised form of academic burnout syndrome. Behind the phenomenon there may be disorders of the hormonal balance, of the hypothalamic-pituitary-adrenal axis, connected with disturbances of dopamine levels (105). Moreover, greater activation of orbitofrontal, anterior cingulate, dorsolateral prefrontal and nucleus accumbens was found when World of Warcraft fans who played more than 30 h per week were exposed to game cues, in comparison to those playing less (106).

Genetic research has pointed out the importance of the CRHR1 gene in gaming addiction (107).

8. Psychopathology

Primary psychiatric disorders that favor addiction must not be confused with psychiatric disorders secondary to the effects of addiction (anxiety, depressive, psychotic disorders, co-addictions).

Based on psychological theories, there are three psychopathological dimensions associated with addictive behaviors: Alexithymia, depression and the search for sensations. Two vulnerability factors appear to be involved: i) Insecure attachments developed in childhood (especially separation anxiety); ii) Depressive vulnerability-the existence of past depressive experiences without being able to objectify a current depression (108,109).

Drug abusers seek anticipated satisfaction from the used substance creating a vicious cycle. Thus, with consumption the satisfaction is obtained, which has an ‘immediate strengthening power’, leading to habit. Meanwhile it accentuates the feeling of incompetence due to the succession of events. Cognitive and behavioral aspects are responsible for strengthening of the habit (110).

An early approach in intrapsychic dynamics, not doubled by the drug control of withdrawal or craving, will increase the impulsivity and the risk of acting out behaviors. An exclusively pharmacological approach can superficially control symptoms without involving profound changes in internal or interrelational dynamics. A double approach is recommended for tackling addiction (111).

9. Conclusion

The present analysis suggests that there are genetic traits behind the development of addiction but internal, behavior and external factors are not to be underestimated. The dopamine-mesolimbic motivation-reward-reinforcement cycle remains the most coherent physiological theory in addiction. The common property of addictive substances is that they are dopamine-agonists; otherwise each class has individual mechanisms, pharmacokinetics and psychoactive potentials, reinforcing the importance of the dopamine-mesolimbic system.

Further knowledge on the genetic, epigenetic, and neurobiological bases of addiction will allow specifically targeted medicine, with higher success rates and lower adverse reactions to compliment the psychological approaches that tackle the behavioral problem in order to diminish this worldwide issue.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

AP and MM designed and drafted the initial review. AMD gathered the medical information and conducted the final view and structure of the article. AP investigated the present area of research and gathered the important information. RVC finalized the work, analysis the results and approved the final version of the work. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ducci F and Goldman D: The genetic basis of addictive disorders. Psychiatr Clin North Am. 35:495–519. 2012.PubMed/NCBI View Article : Google Scholar

2 

American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders. 5th edition. American Psychiatric Association, Washington, DC, 2013.

3 

World Health Organization: ICD-10: International Statistical Classification of Diseases and Related Health Problems: 10th revision, 2nd edition. World Health Organization, 2004. Available from: https://apps.who.int/iris/handle/10665/42980.

4 

Rumpf HJ, Achab S, Billieux J, Bowden-Jones H, Carragher N, Demetrovics Z, Higuchi S, King DL, Mann K, Potenza M, et al: Including gaming disorder in the ICD-11: The need to do so from a clinical and public health perspective. J Behav Addict. 7:556–561. 2018.PubMed/NCBI View Article : Google Scholar

5 

International Statistical Classification of Diseases and Related Health Problems (11th edition) ICD-11, World Health Organization, 2020.

6 

Trifu S, Mihailescu R, Stegarescu S and Ion I: Evolutional Perspective Over Some Key-Aspects in Psychiatry. Theoretical and Applied in Psychology (SICAP23): Psychology and Ongoing Development. Editografica, Bologna, pp179, 2016.

7 

Winger G, Woods JH, Galuska CM and Wade-Galuska T: Behavioral perspectives on the neuroscience of drug addiction. J Exp Anal Behav. 84:667–681. 2005.PubMed/NCBI View Article : Google Scholar

8 

Koob GF and Volkow ND: Neurobiology of addiction: A neurocircuitry analysis. Lancet Psychiatry. 3:760–773. 2016.PubMed/NCBI View Article : Google Scholar

9 

Jacqueline MV: Genetics of Addiction: Future Focus on Gene x environment interaction? J Stud Alcohol Drugs. 77:684–687. 2016.PubMed/NCBI View Article : Google Scholar

10 

Bierut LJ, Madden PA, Breslau N, Johnson EO, Hatsukami D, Pomerleau OF, Swan GE, Rutter J, Bertelsen S, Fox L, et al: Novel genes identified in a high-density genome wide association study for nicotine dependence. Hum Mol Genet. 16:24–35. 2007.PubMed/NCBI View Article : Google Scholar

11 

Dick DM, Viken R, Purcell S, Kaprio J, Pulkkinen L and Rose RJ: Parental monitoring moderates the importance of genetic and environmental influences on adolescent smoking. J Abnorm Psychol. 116:213–218. 2007.PubMed/NCBI View Article : Google Scholar

12 

Hancock DB, Markunas CA, Bierut LJ and Johnson EO: Human genetics of addiction: New insights and future directions. Curr Psychiatry Rep. 20(8)2018.PubMed/NCBI View Article : Google Scholar

13 

Trifu S, Tudor A and Rădulescu I: Aggressive behavior in psychiatric patients in relation to hormonal imbalance (Review). Exp Ther Med. 20:3483–3487. 2020.PubMed/NCBI View Article : Google Scholar

14 

Gräff J and Mansuy IM: Epigenetic dysregulation in cognitive disorders. Eur J Neurosci. 30:1–8. 2009.PubMed/NCBI View Article : Google Scholar

15 

Schmidt HD, McGinty JF, West AE and Sadri-Vakili G: Epigenetics and psychostimulant addiction. Cold Spring Harb Perspect Med. 3(a012047)2013.PubMed/NCBI View Article : Google Scholar

16 

Cadet JL, Brannock C, Jayanthi S and Krasnova IN: Transcriptional and epigenetic substrates of methamphetamine addiction and withdrawal: Evidence from a long-access self-administration model in the rat. Mol Neurobiol. 51:696–717. 2015.PubMed/NCBI View Article : Google Scholar

17 

Cadet JL: Epigenetics of stress, addiction, and resilience: Therapeutic implications. Mol Neurobiol. 53:545–560. 2016.PubMed/NCBI View Article : Google Scholar

18 

Covington HE III, Maze I, Sun H, Bomze HM, DeMaio KD, Wu EY, Dietz DM, Lobo MK, Ghose S, Mouzon E, et al: A role for repressive histone methylation in cocaine-induced vulnerability to stress. Neuron. 71:656–670. 2011.PubMed/NCBI View Article : Google Scholar

19 

Walker CD: Maternal touch and feed as critical regulators of behavioral and stress responses in the offspring. Dev Psychobiol. 52:638–650. 2010.PubMed/NCBI View Article : Google Scholar

20 

Mychasiuk R, Harker A, Ilnytskyy S and Gibb R: Paternal stress prior to conception alters DNA methylation and behaviour of developing rat offspring. Neuroscience. 241:100–105. 2013.PubMed/NCBI View Article : Google Scholar

21 

Benton D and Young HA: A meta-analysis of the relationship between brain dopamine receptors and obesity: A matter of changes in behavior rather than food addiction? Int J Obes (Lond). 40 (Suppl 1):S12–S21. 2016.PubMed/NCBI View Article : Google Scholar

22 

Standaert DG and Walsh RR: Pharmacology of dopaminergic neurotransmission. In: Principles of Pharmacology: The Pathophysiologic Basis of Drug Therapy. Tashjian AH, Armstrong EJ and Golan DE (eds). Lippincott Williams & Wilkins, Philadelphia pp186-206, 2011.

23 

Mishra A, Singh S and Shukla S: Physiological and functional basis of dopamine receptors and their role in neurogenesis: Possible implication for Parkinson's disease. J Exp Neurosci: May 31, 2018 (Epub ahead of print). doi: 10.1177/1179069518779829.

24 

Gardner EL: Addiction and brain reward and antireward pathways. Adv Psychosom Med. 30:22–60. 2011.PubMed/NCBI View Article : Google Scholar

25 

Zhornitsky S, Rizkallah É, Pampoulova T, Chiasson JP, Stip E, Rompré PP and Potvin S: Antipsychotic agents for the treatment of substance use disorders in patients with and without comorbid psychosis. J Clin Psychopharmacol. 30:417–424. 2010.PubMed/NCBI View Article : Google Scholar

26 

Frazer A and Hensler JG: Serotonin Involvement in Physiological Function and Behavior. In: Basic Neurochemistry: Molecular, Cellular and Medical Aspects. Siegel GJ, Agranoff BW and Albers RW (eds). 6th edition. Lippincott-Raven, Philadelphia, 1999.

27 

Palacios JM, Pazos A and Hoyer D: A short history of the 5-HT2C receptor: From the choroid plexus to depression, obesity and addiction treatment. Psychopharmacology (Berl). 234:1395–1418. 2017.PubMed/NCBI View Article : Google Scholar

28 

Maria Alexandra Stanescu A, Totan A, Mircescu D, Diaconescu S, Gabriel Bratu O, Fekete L, László Fekete G, Boda D and Cristina Diaconu C: Assessment of suicidal behavior in dermatology (Review). Exp Ther Med. 20:73–77. 2020.PubMed/NCBI View Article : Google Scholar

29 

Kirby LG, Zeeb FD and Winstanley CA: Contributions of serotonin in addiction vulnerability. Neuropharmacology. 61:421–432. 2011.PubMed/NCBI View Article : Google Scholar

30 

Vogeler T, McClain C and Evoy KE: Combination bupropion SR and varenicline for smoking cessation: A systematic review. Am J Drug Alcohol Abuse. 42:129–139. 2016.PubMed/NCBI View Article : Google Scholar

31 

Zagon IS and McLaughlin PJ: Endogenous Opioids in the Etiology and Treatment of Multiple Sclerosis. In: Multiple Sclerosis: Perspectives in Treatment and Pathogenesis. Zagon IS and McLaughlin PJ (eds). Codon Publications, Brisbane, AU, 2017.

32 

Herman JP, Figueiredo HF, Mueller NK, Ostrander MM, Zhang R, Tauchi M, Choi DC, Furay AR, Evanson NK, Nelson EB, et al: Neurochemical Systems Regulating the Hypothalamo-Pituitary-Adrenocortical Axis. In: Handbook of Neurochemistry and Molecular Neurobiology. Lajtha A and Blaustein JD (eds). Springer, Boston, pp513-569, 2007.

33 

Browning KN and Travagli RA: Central nervous system control of gastrointestinal motility and secretion and modulation of gastrointestinal functions. Compr Physiol. 4:1339–1368. 2014.PubMed/NCBI View Article : Google Scholar

34 

Gilpin NW and Koob GF: Neurobiology of alcohol dependence: Focus on motivational mechanisms. Alcohol Res Health. 31:185–195. 2008.PubMed/NCBI

35 

Aboujaoude E and Salame WO: Naltrexone: A Pan-Addiction treatment? CNS Drugs. 30:719–733. 2016.PubMed/NCBI View Article : Google Scholar

36 

Mark GP, Shabani S, Dobbs LK and Hansen ST: Cholinergic modulation of mesolimbic dopamine function and reward. Physiol Behav. 104:76–81. 2011.PubMed/NCBI View Article : Google Scholar

37 

Sofuoglu M and Mooney M: Cholinergic functioning in stimulant addiction: Implications for medications development. CNS Drugs. 23:939–952. 2009.PubMed/NCBI View Article : Google Scholar

38 

Williams MJ and Adinoff B: The role of acetylcholine in cocaine addiction. Neuropsychopharmacology. 33:1779–1797. 2008.PubMed/NCBI View Article : Google Scholar

39 

Enoch MA: The role of GABA(A) receptors in the development of alcoholism. Pharmacol Biochem Behav. 90:95–104. 2008.PubMed/NCBI View Article : Google Scholar

40 

Tyacke RJ, Lingford-Hughes A, Reed LJ and Nutt DJ: GABAB receptors in addiction and its treatment. Adv Pharmacol. 58:373–396. 2010.PubMed/NCBI View Article : Google Scholar

41 

Minozzi S, Saulle R and Rösner S: Baclofen for alcohol use disorder. Cochrane Database Syst Rev. 11(CD012557)2018.PubMed/NCBI View Article : Google Scholar

42 

Volkow ND and Morales M: The Brain on drugs: From reward to addiction. Cell. 162:712–725. 2015.PubMed/NCBI View Article : Google Scholar

43 

Li CT, Yang KC and Lin WCL: Glutamatergic dysfunction and glutamatergic compounds for major psychiatric disorders: Evidence from clinical neuroimaging studies. Front Psychiatry. 9(767)2019.PubMed/NCBI View Article : Google Scholar

44 

Schultz W: Neuronal reward and decision signals: From theories to data. Physiol Rev. 95:853–951. 2015.PubMed/NCBI View Article : Google Scholar

45 

Cooper S, Robison AJ and Mazei-Robison MS: Reward circuitry in addiction. Neurotherapeutics. 14:687–697. 2017.PubMed/NCBI View Article : Google Scholar

46 

Stott SRW and Ang SL: The Generation of midbrain dopaminergic neurons. In: Patterning and Cell Type Specification in the Developing CNS and PNS. Rubenstein JLR and Rakic P (eds). Academic Press, Cambridge, pp435-453, 2013.

47 

Olds J and Milner P: Positive reinforcement produced by electrical stimulation of the septal area and other regions of rat brain. J Comp Physiol Psychol. 47:419–27. 1954.PubMed/NCBI View Article : Google Scholar

48 

Goeders NE and Smith JE: Cortical dopaminergic involvement in cocaine reinforcement. Science. 221:773–775. 1983.PubMed/NCBI View Article : Google Scholar

49 

Edenberg H and Foroud T: Genetics and alcoholism. Nat Rev Gastroenterol Hepatol. 10:487–494. 2013.PubMed/NCBI View Article : Google Scholar

50 

Vetreno RP and Crews FT: Current hypotheses on the mechanisms of alcoholism. Handb Clin Neurol. 125:477–497. 2014.PubMed/NCBI View Article : Google Scholar

51 

Sullivan EV and Pfefferbaum A: Neurocircuitry in alcoholism: A substrate of disruption and repair. Psychopharmacology (Berl). 180:583–594. 2005.PubMed/NCBI View Article : Google Scholar

52 

Sharp BM and Chen H: Neurogenetic determinants and mechanisms of addiction to nicotine and smoked tobacco. Eur J Neurosci. 50:2164–2179. 2019.PubMed/NCBI View Article : Google Scholar

53 

Benowitz NL: Pharmacology of nicotine: Addiction, smoking-induced disease and therapeutics. Annu Rev Pharmacol Toxicol. 49:57–71. 2009.PubMed/NCBI View Article : Google Scholar

54 

D'Souza MS and Markou A: Neuronal mechanisms underlying development of nicotine dependence: Implications for novel smoking-cessation treatments. Addict Sci Clin Pract. 6:4–16. 2011.PubMed/NCBI

55 

Durazzo TC, Meyerhoff DJ and Nixon SJ: Chronic cigarette smoking: Implications for neurocognition and brain neurobiology. Int J Environ Res Public Health. 7:3760–3791. 2010.PubMed/NCBI View Article : Google Scholar

56 

Nichols DE: Psychedelics. Pharmacol Rev. 68:264–355. 2016.PubMed/NCBI View Article : Google Scholar

57 

López-Giménez JF and González-Maeso J: Hallucinogens and Serotonin 5-HT(2A) receptor-mediated signaling pathways. Curr Top Behav Neurosci. 36:45–73. 2018.PubMed/NCBI View Article : Google Scholar

58 

Weir E: Inhalant use and addiction in Canada. CMAJ. 164(397)2001.PubMed/NCBI

59 

Kumar S, Grover S, Kulhara P, Mattoo SK, Basu D, Biswas P and Shah R: Inhalant abuse: A clinic-based study. Indian J Psychiatry. 50:117–120. 2008.PubMed/NCBI View Article : Google Scholar

60 

Bloomfield MAP, Hindocha C, Green SF, Wall MB, Lees R, Petrilli K, Costello H, Ogunbiyi MO, Bossong MG and Freeman TP: The neuropsychopharmacology of cannabis: A review of human imaging studies. Pharmacol Ther. 195:132–161. 2019.PubMed/NCBI View Article : Google Scholar

61 

Devinsky O, Patel AD, Cross JH, Villanueva V, Wirrell EC, Privitera M, Greenwood SM, Roberts C, Checketts D, VanLandingham KE, et al: Effect of cannabidiol on drop seizures in the Lennox-Gastaut syndrome. N Engl J Med. 378:1888–1897. 2018.PubMed/NCBI View Article : Google Scholar

62 

McGuire P, Robson P, Cubala WJ, Vasile D, Morrison PD, Barron R, Taylor A and Wright S: Cannabidiol (CBD) as an adjunctive therapy in schizophrenia: A multicenter randomized controlled trial. Am J Psychiatry. 175:225–231. 2018.PubMed/NCBI View Article : Google Scholar

63 

Wijayendran SB, O'Neill A and Bhattacharyya S: The effects of cannabis use on salience attribution: A systematic review. Acta Neuropsychiatr. 30:43–57. 2018.PubMed/NCBI View Article : Google Scholar

64 

Colizzi M, McGuire P, Pertwee RG and Bhattacharyya S: Effect of cannabis on glutamate signalling in the brain: A systematic review of human and animal evidence. Neurosci Biobehav Rev. 64:359–381. 2016.PubMed/NCBI View Article : Google Scholar

65 

Hirvonen J, Goodwin RS, Li CT, Terry GE, Zoghbi SS, Morse C, Pike VW, Volkow ND, Huestis MA and Innis RB: Reversible and regionally selective downregulation of brain cannabinoid CB1 receptors in chronic daily cannabis smokers. Mol Psychiatry. 17:642–649. 2017.PubMed/NCBI View Article : Google Scholar

66 

Renard J, Vitalis T, Rame M, Krebs MO, Lenkei Z, Le Pen G and Jay TM: Chronic cannabinoid exposure during adolescence leads to long-term structural and functional changes in the prefrontal cortex. Eur Neuropsychopharmacol. 26:55–64. 2016.PubMed/NCBI View Article : Google Scholar

67 

Saffroy R, Lafaye G, Desterke C, Ortiz-Tudela E, Amirouche A, Innominato P, Pham P, Benyamina A and Lemoine A: Several clock genes polymorphisms are meaningful risk factors in the development and severity of cannabis addiction. Chronobiol Int. 36:122–134. 2019.PubMed/NCBI View Article : Google Scholar

68 

Gerra MC, Manfredini M, Cortese E, Antonioni MC, Leonardi C, Magnelli F, Somaini L, Jayanthi S, Cadet JL and Donnini C: Genetic and environmental risk factors for cannabis use: Preliminary results for the role of parental care perception. Subst Use Misuse. 54:670–680. 2019.PubMed/NCBI View Article : Google Scholar

69 

Hodgson K, Coleman JR, Hagenaars SP, Purves KL, Glanville K, Choi SW, O'Reilly P and Breen G: Major Depressive Disorder Working Group of the Psychiatric Genomics Consortium. Lewis CM: Cannabis use, depression and self-harm: Phenotypic and genetic relationships. Addiction. 115:482–492. 2020.PubMed/NCBI View Article : Google Scholar

70 

Montalvo-Ortiz JL, Zhou H, D'Andrea I, Maroteaux L, Lori A, Smith A, Ressler KJ, Nuñez YZ, Farrer LA, Zhao H, et al: Translational studies support a role for serotonin 2B receptor (HTR2B) gene in aggression-related cannabis response. Mol Psychiatry. 23:2277–2286. 2018.PubMed/NCBI View Article : Google Scholar

71 

Pasman JA, Verweij KJ, Gerring Z, Stringer S, Sanchez-Roige S, Treur JL, Abdellaoui A, Nivard MG, Baselmans BM, Ong JS, et al: GWAS of lifetime cannabis use reveals new risk loci, genetic overlap with psychiatric traits, and a causal influence of schizophrenia. Nat Neurosci. 21:1161–1170. 2018.PubMed/NCBI View Article : Google Scholar

72 

Petrovska J, Coynel D, Fastenrath M, Milnik A, Auschra B, Egli T, Gschwind L, Hartmann F, Loos E, Sifalakis K, et al: The NCAM1 gene set is linked to depressive symptoms and their brain structural correlates in healthy individuals. J Psychiatr Res. 91:116–123. 2017.PubMed/NCBI View Article : Google Scholar

73 

Trifu S, Vladuți A and Trifu AI: Genetic aspects in schizophrenia. Receptoral theories. Metabolic theories. Rom J Morphol Embryol. 61:25–32. 2020.PubMed/NCBI View Article : Google Scholar

74 

Spahn V, Fischer O, Endres-Becker J, Schäfer M, Stein C and Zöllner C: Opioid withdrawal increases transient receptor potential vanilloid 1 activity in a protein kinase A-dependent manner. Pain. 154:598–608. 2013.PubMed/NCBI View Article : Google Scholar

75 

Le Merrer J, Becker JA, Befort K and Kieffer BL: Reward processing by the opioid system in the brain. Physiol Rev. 89:1379–412. 2009.PubMed/NCBI View Article : Google Scholar

76 

Schramm-Sapyta NL, Walker QD, Caster JM, Levin ED and Kuhn CM: Are adolescents more vulnerable to drug addiction than adults? Evidence from animal models. Psychopharmacology (Berl). 206:1–21. 2009.PubMed/NCBI View Article : Google Scholar

77 

Van'tVeer A, Yano JM, Carroll FI, Cohen BM and Carlezon WA Jr: Corticotropin-releasing factor (CRF)-induced disruption of attention in rats is blocked by the κ-opioid receptor antagonist JDTic. Neuropsychopharmacology. 37:2809–2816. 2012.PubMed/NCBI View Article : Google Scholar

78 

Wang S: Historical review: Opiate addiction and opioid receptors. Cell Transplant. 28:233–238. 2019.PubMed/NCBI View Article : Google Scholar

79 

Wang SC, Chen YC, Lee CH and Cheng CM: Opioid addiction, genetic susceptibility and medical treatments: A review. Int J Mol Sci. 20(4294)2019.PubMed/NCBI View Article : Google Scholar

80 

Gelernter J, Kranzler HR, Sherva R, Koesterer R, Almasy L, Zhao H and Farrer LA: Genome-wide association study of opioid dependence: Multiple associations mapped to calcium and potassium pathways. Biol Psychiatry. 76:66–74. 2014.PubMed/NCBI View Article : Google Scholar

81 

Kreek MJ, Nielsen DA and LaForge KS: Genes associated with addiction: Alcoholism, opiate, and cocaine addiction. Neuromolecular Med. 5:85–108. 2004.PubMed/NCBI View Article : Google Scholar

82 

Brett J and Murnion B: Management of benzodiazepine misuse and dependence. Aust Prescr. 38:152–155. 2015.PubMed/NCBI View Article : Google Scholar

83 

Blum K, Gold M, Modestino EJ, Baron D, Boyett B, Siwicki D, Lott L, Podesta A, Roy AK, Hauser M, et al: Would induction of dopamine homeostasis via coupling genetic addiction risk score (GARS®) and pro-dopamine regulation benefit benzodiazepine use disorder (BUD)? J Syst Integr Neurosci: May 3, 2018 (Epub ahead of print). doi: 10.15761/JSIN.1000196.

84 

Blum K, Modestino EJ, Lewis MG, Baron D, Steinberg B, Thanos PK, Downs BW, Siwicki D, Lott L, Braverman E, et al: Pro-dopamine regulator (KB220) a fifty year sojourn to combat reward deficiency syndrome (RDS): Evidence based bibliography (Annotated). CPQ Neurol Psychol: Dec 4, 2018 (Epub ahead of print).

85 

Konopka A, Mak M, Grzywacz A, Murawiec S and Samochowiec J: Genetic and psychosocial factors for benzodiazepine addiction. An analysis based on the results of the authors' own research conducted in a group of benzodiazepines addicted and non-addicted individuals. Postepy Hig Med Dosw (Online). 71:205–213. 2017.PubMed/NCBI View Article : Google Scholar

86 

Gossop M, Griffiths P, Powis B and Strang J: Cocaine: Patterns of use, route of administration, and severity of dependence. Br J Psychiatry. 164:660–664. 1994.PubMed/NCBI View Article : Google Scholar

87 

Coe MA, Jufer Phipps RA, Cone EJ and Walsh SL: Bioavailability and pharmacokinetics of oral cocaine in humans. J Anal Toxicol. 42:285–292. 2018.PubMed/NCBI View Article : Google Scholar

88 

Olivares EL, Kendler KS, Neale MC and Gillespie NA: The genetic and environmental association between parental monitoring and risk of cannabis, stimulants, and cocaine initiation in a sample of male twins: Does parenting matter? Twin Res Hum Genet. 19:297–305. 2016.PubMed/NCBI View Article : Google Scholar

89 

Jensen KP: A review of genome-wide association studies of stimulant and opioid use disorders. Mol Neuropsychiatry. 2:37–45. 2016.PubMed/NCBI View Article : Google Scholar

90 

Le Q, Yan B, Yu X, Li Y, Song H, Zhu H, Hou W, Ma D, Wu F, Zhou Y and Ma L: Drug-seeking motivation level in male rats determines offspring susceptibility or resistance to cocaine-seeking behaviour. Nat Commun. 8(15527)2017.PubMed/NCBI View Article : Google Scholar

91 

Sepanjnia K, Modabbernia A, Ashrafi M, Modabbernia MJ and Akhondzadeh S: Pioglitazone adjunctive therapy for moderate-to-severe major depressive disorder: Randomized double-blind placebo-controlled trial. Neuropsychopharmacology. 37:2093–2100. 2012.PubMed/NCBI View Article : Google Scholar

92 

Chen X, Xue L, Hou S, Jin Z, Zhang T, Zheng F and Zhan CG: Long-acting cocaine hydrolase for addiction therapy. Proc Natl Acad Sci USA. 113:422–427. 2016.PubMed/NCBI View Article : Google Scholar

93 

Yu S, Zhu L, Shen Q, Bai X and Di X: Recent advances in methamphetamine neurotoxicity mechanisms and its molecular pathophysiology. Behav Neurol. 2015(103969)2015.PubMed/NCBI View Article : Google Scholar

94 

LaVoie MJ, Card JP and Hastings TG: Microglial activation precedes dopamine terminal pathology in methamphetamine-induced neurotoxicity. Exp Neurol. 187:47–57. 2004.PubMed/NCBI View Article : Google Scholar

95 

Cadet JL and Bisagno V: Glial-neuronal ensembles: Partners in drug addiction-associated synaptic plasticity. Front Pharmacol. 5(204)2014.PubMed/NCBI View Article : Google Scholar

96 

Krasnova IN, Justinova Z and Cadet JL: Methamphetamine addiction: Involvement of CREB and neuroinflammatory signaling pathways. Psychopharmacology (Berl). 233:1945–1962. 2016.PubMed/NCBI View Article : Google Scholar

97 

Olsen CM: Natural rewards, neuroplasticity, and non-drug addictions. Neuropharmacology. 61:1109–1122. 2011.PubMed/NCBI View Article : Google Scholar

98 

Lobo DS, Souza RP, Tong RP, Casey DM, Hodgins DC, Smith GJ, Williams RJ, Schopflocher DP, Wood RT, el-Guebaly N and Kennedy JL: Association of functional variants in the dopamine D2-like receptors with risk for gambling behaviour in healthy Caucasian subjects. Biol Psychol. 85:33–7. 2010.PubMed/NCBI View Article : Google Scholar

99 

Yau YH and Potenza MN: Gambling disorder and other behavioral addictions: Recognition and treatment. Harv Rev Psychiatry. 23:134–146. 2015.PubMed/NCBI View Article : Google Scholar

100 

Moragas L, Granero R, Stinchfield R, Fernández-Aranda F, Fröberg F, Aymamí N, Gómez-Peña M, Fagundo AB, Islam MA, Del Pino-Gutiérrez A, et al: Comparative analysis of distinct phenotypes in gambling disorder based on gambling preferences. BMC Psychiatry. 15(86)2015.PubMed/NCBI View Article : Google Scholar

101 

Kessler RC, Hwang I, LaBrie R, Petukhova M, Sampson NA, Winters KC and Shaffer HJ: DSM-IV pathological gambling in the National comorbidity survey replication. Psychol Med. 38:1351–60. 2008.PubMed/NCBI View Article : Google Scholar

102 

Grant JE, Odlaug BL and Chamberlain SR: Neural and psychological underpinnings of gambling disorder: A review. Prog Neuropsychopharmacol Biol Psychiatry. 65:188–193. 2016.PubMed/NCBI View Article : Google Scholar

103 

Wilson D, da Silva Lobo DS, Tavares H, Gentil V and Vallada H: Family-based association analysis of serotonin genes in pathological gambling disorder: Evidence of vulnerability risk in the 5HT-2A receptor gene. J Mol Neurosci. 49:550–553. 2013.PubMed/NCBI View Article : Google Scholar

104 

Weinstein AM: Computer and video game addiction-a comparison between game users and non-game users. Am J Drug Alcohol Abuse. 36:268–276. 2010.PubMed/NCBI View Article : Google Scholar

105 

Trifu S: Neuroendocrine insights into burnout syndrome. Acta Endocrinol (Buchar). 15:404–405. 2019.PubMed/NCBI View Article : Google Scholar

106 

Ko CH, Liu GC, Hsiao S, Yen JY, Yang MJ, Lin WC, Yen CF and Chen CS: Brain activities associated with gaming urge of online gaming addiction. J Psychiatr Res. 43:739–747. 2009.PubMed/NCBI View Article : Google Scholar

107 

Park J, Sung JY, Kim DK, Kong ID, Hughes TL and Kim N: Genetic association of human Corticotropin-releasing hormone receptor 1 (CRHR1) with Internet gaming addiction in Korean male adolescents. BMC Psychiatry. 18(396)2018.PubMed/NCBI View Article : Google Scholar

108 

Franchina V, Vanden Abeele M, Van Rooij AJ, Lo Coco G and De Marez L: Fear of Missing out as a predictor of problematic social media use and phubbing behavior among Flemish adolescents. Int J Environ Res Public Health. 15(2319)2018.PubMed/NCBI View Article : Google Scholar

109 

Trifu S and Gutt A: Interpretative process-from utilization of predominant to psychotic decompensation. Procedia Soc Behav Sci. 187:429–433. 2014.

110 

Ostlund SB and Balleine BW: On habits and addiction: An associative analysis of compulsive drug seeking. Drug Discov Today Dis Models. 5:235–245. 2008.PubMed/NCBI View Article : Google Scholar

111 

Powell BJ, McMillen JC, Proctor EK, Carpenter CR, Griffey RT, Bunger AC, Glass JE and York JL: A compilation of strategies for implementing clinical innovations in health and mental health. Med Care Res Rev. 69:123–157. 2012.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

May-2021
Volume 21 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Popescu A, Marian M, Drăgoi AM and Costea R: Understanding the genetics and neurobiological pathways behind addiction (Review). Exp Ther Med 21: 544, 2021
APA
Popescu, A., Marian, M., Drăgoi, A.M., & Costea, R. (2021). Understanding the genetics and neurobiological pathways behind addiction (Review). Experimental and Therapeutic Medicine, 21, 544. https://doi.org/10.3892/etm.2021.9976
MLA
Popescu, A., Marian, M., Drăgoi, A. M., Costea, R."Understanding the genetics and neurobiological pathways behind addiction (Review)". Experimental and Therapeutic Medicine 21.5 (2021): 544.
Chicago
Popescu, A., Marian, M., Drăgoi, A. M., Costea, R."Understanding the genetics and neurobiological pathways behind addiction (Review)". Experimental and Therapeutic Medicine 21, no. 5 (2021): 544. https://doi.org/10.3892/etm.2021.9976