Next Article in Journal
FBXL7 Body Hypomethylation Is Frequent in Tumors from the Digestive and Respiratory Tracts and Is Associated with Risk-Factor Exposure
Next Article in Special Issue
In Vivo Bio-Activation of JWH-175 to JWH-018: Pharmacodynamic and Pharmacokinetic Studies in Mice
Previous Article in Journal
Superhydrophobic Paper-Based Microfluidic Field-Effect Transistor Biosensor Functionalized with Semiconducting Single-Walled Carbon Nanotube and DNAzyme for Hypocalcemia Diagnosis
Previous Article in Special Issue
Amitriptyline Accelerates SERT Binding Recovery in a Rat 3,4-Methylenedioxymethamphetamine (MDMA) Model: In Vivo 4-[18F]-ADAM PET Imaging
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ketamine plus Alcohol: What We Know and What We Can Expect about This

by
Natalia Harumi Correa Kobayashi
1,†,
Sarah Viana Farias
1,†,
Diandra Araújo Luz
1,
Kissila Márvia Machado-Ferraro
1,
Brenda Costa da Conceição
1,
Cinthia Cristina Menezes da Silveira
1,
Luanna Melo Pereira Fernandes
1,
Sabrina de Carvalho Cartágenes
1,
Vânia Maria Moraes Ferreira
2,
Enéas Andrade Fontes-Júnior
1 and
Cristiane do Socorro Ferraz Maia
1,*
1
Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075110, PA, Brazil
2
Laboratory of Psychobiology, Psychology Institute, University of Brasília, Campus Universitário Darcy Ribeiro—Asa Norte, Brasília 70910900, DF, Brazil
*
Author to whom correspondence should be addressed.
These authors contributed equally to the work.
Int. J. Mol. Sci. 2022, 23(14), 7800; https://doi.org/10.3390/ijms23147800
Submission received: 4 June 2022 / Revised: 23 June 2022 / Accepted: 27 June 2022 / Published: 15 July 2022
(This article belongs to the Special Issue Pharmaco-Toxicological Effects of Novel Psychoactive Substances 2.0)

Abstract

:
Drug abuse has become a public health concern. The misuse of ketamine, a psychedelic substance, has increased worldwide. In addition, the co-abuse with alcohol is frequently identified among misusers. Considering that ketamine and alcohol share several pharmacological targets, we hypothesize that the consumption of both psychoactive substances may synergically intensify the toxicological consequences, both under the effect of drugs available in body systems and during withdrawal. The aim of this review is to examine the toxicological mechanisms related to ketamine plus ethanol co-abuse, as well the consequences on cardiorespiratory, digestive, urinary, and central nervous systems. Furthermore, we provide a comprehensive discussion about the probable sites of shared molecular mechanisms that may elicit additional hazardous effects. Finally, we highlight the gaps of knowledge in this area, which deserves further research.

1. Introduction

Drug abuse is an ancient human practice, which involves genetic and environmental factors, as well as a great complexity of neuronal circuitry [1]. Currently, the use of psychoactive substances has become a worldwide concern and public health issue, due to health risks and social problems [2,3]. In general, such involvement in drug misuse occurs early in life. On a global scale, it is estimated that 1 among 20 individuals, ranging from 15 to 64 years old, have already consumed any recreational substance to achieve altered states of consciousness [4,5].
Of note, ethanol consists of the most widely consumed psychoactive substance [6]. According to the World Health Organization (2014), the harmful use of ethanol is responsible for about 5.9% of deaths in the world, as a consequence of traffic accidents, social interaction problems, domestic violence, crimes, public disorder, and chronic health problems [7,8,9]. Moreover, the consumption of alcohol is a factor that can lead to the consumption of additional psychoactive drugs, such as methylenedioxymethamphetamine (MDMA), methamphetamine, lysergic acid diethylamide (LSD), gamma-hydroxybutyrate (GHB), and especially ketamine, which has been in evidence among drug abusers [10].
Ketamine has been used for non-therapeutic purposes, especially as a “club drug”, being referred to as “Key”, “Special K”, “angel dust”, “K”, or “Kit Kat” [11]. Ketamine is commonly misused by intranasal route, inhaled, or smoked [12]. When administered in liquid form, users inject it into the body or mix it in alcoholic drinks for oral route [13].
It is noteworthy that both psychoactive drugs share several mechanisms of action on the central nervous system (CNS). Primarily, ethanol evokes non-competitive inhibition of N-methyl-D-aspartate (NMDA) receptors [14,15,16]. Ketamine also acts through an NMDA open-channel blockade mechanism [17,18]. Although both drugs act equally on the same receptor, it is unclear whether ketamine can modify the molecular effects of alcohol on NMDA receptor [14,19]. Thus, we ask whether co-exposure to these substances could imply a synergistic toxicological effect mediated by the NMDA pathway, intensifying toxicological responses.
Several and robust preclinical and clinical studies have described the hazardous effects elicited by alcohol consumption, even after long-lasting withdrawal [20,21,22,23]. On the other side, ketamine non-medical purpose use has been described in only a few studies in the last decades [12,24,25,26]. Unfortunately, studies that have researched the consequences of ethanol plus ketamine concomitant exposure in body systems are scarce. Thus, we decided to collect available clinical and preclinical studies related to the recreational use of ketamine plus ethanol, focusing on its detrimental consequences. In addition, we propose the probable mechanism of the toxicological synergic effects that might contribute to hazardous effects of this co-consumption.

2. Epidemiological Features of Ethanol plus Ketamine Consumption

According to the 2018 Global Report on Alcohol and Health published by the World Health Organization (WHO), young adults, ranging from 20 to 39 years old, are the main consumers of alcohol [7]. In 2010, a survey reported that male alcohol intake was higher than that of females, with a consumption of around 19.4 L/year. In contrast, women consume about 7 L/year [27], but such a pattern of global alcohol consumption has been modified, since women have augmented alcohol drinking over the years in amounts that are increasingly closer to men [28]. Of particular interest is the co-ingestion of alcohol with other drugs by abusers [2,10,13].
Ketamine consists of a dissociative anesthetic applied to sedation and analgesia procedures and some psychiatry conditions [29,30,31]. Although there are few epidemiological data on the non-medical use of ketamine, it has been reported that ketamine misuse started in the 1970s [32]. It is used primarily in the United States, but nowadays the recreational use has spread worldwide [33]. Thus, ketamine is included as a new psychoactive substance (NPS) in drug-abuse criteria [11,31,34]. Currently, the Asian continent is emerging as a major geographic region that consumes this substance; ketamine is ranked as the first choice for misused drug among young abusers in Hong Kong. In Western countries, ketamine misuse is around 1–2% [35]. However, the recreational use of this drug has also increased in United Kingdom, Australia, and China [13]. In a French survey between 2012 and 2017, ketamine misuse was prevalent in 67% of NPSs detected [34]. From this information, we suggest that ketamine recreational use is not restricted to some countries; on the contrary, such psychoactive substance is overspread worldwide.
The concomitant use of ethanol plus ketamine has been described. In an emergency department of Bologna (Italy), alcohol was present with 25% of ketamine recreational misusers admitted to emergency care [36]. In accordance, the abuser’s co-ingestion percentage was around 39–98% in the preceding 12 months [37,38]. Unfortunately, both studies failed to link the negative symptoms to the possible synergism of toxicological effects of both drugs users were exposed to. However, a more prevalent association was found in controlled studies with volunteers recruited at party scenes, of which at least 65% of ketamine users also consume alcohol [24,39,40]. A wide survey reported that alcohol is present at a percentual of 98% of ketamine abusers, in a co-administration pattern or not [38]. Actually, all of these findings suggest the close relationship between ketamine and alcohol consumption.

Fatal Outcomes

A positive relationship between recreational drugs plus alcohol and fatal or non-fatal overdose has been postulated [41,42,43].
In a wide Australian survey (2000–2019), ketamine self-administration as contributory to death was investigated [44]. In Darke and colleagues’ study [44], ethanol was present in a over a quarter (27.3%) of ketamine-related deaths. Previously, in the United Kingdom (1993–2006), alcohol was present in almost 50% of postmortem analyses from ketamine-related deaths [45]. Such few reports highlight the occurrence of polydrug use among addicts, in which alcohol consumption is prevalent. Of note, the limitation of the studies that clearly establish the contribution of each drug to a fatal outcome is obvious, since ketamine and alcohol are commonly associated with the use of other psychoactive drugs [44].

3. Body Systems Consequences of Ketamine plus Ethanol Abuse

3.1. Liver and Biliary System Damage

The recreational use of ketamine has been reported as a factor for eliciting abdominal pains related to unknown etiology (for a review, see Reference [46]). Such characteristic symptoms are colloquially referred to as “K-cramps” [47] and consist of several claims reported by ketamine abusers in emergency departments, with the occurrence of dilated common bile duct and choledochal cysts [48,49,50]. In addition, abnormal liver function and gallbladder dyskinesia have been found in clinical studies of ketamine abusers [48,49,51,52,53]. The exact mechanism that underlies ketamine hepatotoxicity is unknown; however, a clinical study has postulated that a direct toxic effect on parenchymal hepatic cells might occur [54]. Actually, an in vitro study revealed that ketamine induces hepatotoxicity through apoptotic mechanisms, specially reducing the mitochondrial membrane potential and ATP synthesis [55]. In addition, it is speculated that ketamine directly blocks the NMDA receptor and calcium channels’ smooth muscle cells of the biliary system; this may contribute to biliary dilatation [51]. The indirect mechanism of ketamine’s hepatobiliary hazardous effects relies on the inhibition of the dorsal motor nucleus of vagus through the NMDA blockade; this also leads to gallbladder dyskinesia [56]. All of the molecular hazardous effects mentioned above are related to ketamine’s pharmacological mechanism, which requires plasmatic concentration of this substance. Finally, ketamine’s pharmacokinetic profiles consist of a wide hepatic CYP P-450 inducer, which can trigger the toxicity of other drugs in the hepatobiliary system or other organs, through elevated production of toxic metabolites, which may last even during withdrawal [55].
On the other hand, the detrimental relationship between alcohol and the liver has been extensively reported (for a review, see Reference [57]). There are three main pathogenic mechanisms related to alcohol ingestion in the liver: oxidative damage, pro-inflammatory processes, and hepatocellular alterations elicited by ethanol and its metabolites [58]. In fact, there are direct hepatotoxic consequences of alcohol metabolites, mainly acetaldehyde, acetate, and fatty acid ethanol esters (FAEEs) [58]. Acetaldehyde and its subproduct, acetate, have a crucial role in alcohol-induced liver disease (ALD). Such metabolites are the products that result from the oxidation of alcohol by the cytoplasmic alcohol dehydrogenase (ALDH) enzyme, which, in general, is overexpressed in chronic ethanol consumption, followed by acetaldehyde dehydrogenase (ALDH), respectively, inducing toxicological liver effects and displaying hepatic fibrosis by a harmful feedback cycle, i.e., detrimental extra-cellular matrix remodeling, oxidative stress, and pro-inflammatory events [58,59]. All of these toxicological events occur as a result of blood alcohol concentration; however, they can persist during long-term withdrawal [23].
Although the epidemiological ketamine abuse surveys have not cleared the contribution of alcohol on gastrointestinal symptoms, such reports have highlighted that the co-abuse with alcohol has been prevalent (around 30%) [36,48,60]. Considering the findings above, we suggest that the co-abuse of ketamine plus ethanol by misusers may elicit cirrhosis, as well as increase collagen fibers in the liver [61,62] (Figure 1B). Actually, an experimental study revealed that high levels of cell death via a necrosis process have been detected in the livers of rats that received a ketamine-plus-ethanol regiment [61]. In addition, solely a case-report study has described the hepatobiliary symptoms observed in ethanol plus ketamine co-abuse [63], thus highlighting the non-relevance/non-importance of such co-abuse among clinicians.

3.2. Cardiorespiratory System

Epidemiological findings have reported the cardiovascular and respiratory systems’ consequences in the emergency services related to ketamine recreational use [48,60].
Palpitations, tachycardia, chest pain, and hypertension are symptoms claimed by ketamine abusers in the emergency department; these symptoms are related to cardiovascular toxicity [46,53,60,64], which may be generated through the hyperactivation of the reflex sympathetic [64,65]. Actually, ketamine presents pleiotropic effects, in which the inhibition of cholinergic transmission, as well as the blockade of neuronal and non-neuronal reuptake of norepinephrine, induces a persistent adrenergic systemic response [65]. Indeed, oxidative stress may play an important role in the cardiotoxicity induced by ketamine [66]. These effects are related to the pharmacological mechanisms of ketamine bioavailability, in which oxidative unbalance might persist.
On the other side, the involvement of alcohol on cardiorespiratory system is well established [67]. Acute or chronic ethanol consumption and its metabolites in systemic circulation (i.e., acetaldehyde) elicit arrhythmias and cardiomyopathy [67]. Negative inotropic effects, cardiac contractile dysfunction, myofibrillar structure alteration, and prolonged cardiac repolarization (QT interval) effects resulting from calcium/calmodulin-dependent protein kinase II (CaMKII) signaling hyperactivation, reactive oxygen species (ROS) overproduction, and cardiac ions homeostasis disturbance lead to cardiac failure [67].
Such cardiac toxicological overlapping may explain the increase of the toxicological evidence found in the co-ingested drugs [64]. Figure 1C shows the probable synergic toxicological mechanisms related to ketamine plus ethanol consumption on heart injury.
In the respiratory system, apnea, pulmonary edema, and respiratory depression have been observed in humans after ketamine misuse, as a result of its pleiotropic pharmacological actions [46,64]. In neurotransmitter systems, ketamine increases serotonin, norepinephrine, opioid, and GABAergic pathways’ activation [65], affecting ventilatory responses, which depend on the neurons or receptors activated (i.e., α2-adrenergic receptor), that can display depression of respiratory motor activity [68]. Ketamine is a weak agonist of the mu-opioid and GABAa receptors; however, at high doses, it increases GABA signaling [65]. It is well documented that mu-opioid and GABA receptors are involved in respiratory depression [46,69]. In addition, NMDA receptors plays a pivotal role in regard to ventilatory responses, in which the blockade of NMDA receptors also contributes to respiratory depression [70]. We suggest that synergic mechanisms of the multiple sites of ketamine action may induce respiratory system depression when higher plasmatic concentrations are reached [65].
In addition to the above effects, ethanol exposure also predisposes to lung injury (i.e., bronchitis, pulmonary edema, fibrosis, and pneumonia), primarily through oxidative damage [71]. Reduction of pulmonary innate defense mechanisms and adaptative immune responses have been displayed by mucociliary clearance dysfunction, impaired alveolar macrophages phagocytosis, decreased CD4+/CD8+-T cells and interferon-γ (IFN-γ) mediator, and breakdown of the protective cytokine signaling responses (for a review, see Reference [71]). Epithelial permeability of the respiratory airway is affected by ethanol, inducing pulmonary oedema through ion (Na+, Cl, and K+) transport dysfunction, disturbance of claudin protein homeostasis, eliciting weakness of tight junction and pulmonary barrier [71]. Moreover, the increase of extracellular matrix deposition and overactivation of transforming growth factor beta 1 (TGF-β1) and metalloproteinases are responsible for fibrosis in the lung [71]. Such accessory detrimental effects emerge as a consequence of alcohol abuse in a long-term period in withdrawal.
As explained in regard to the cardiotoxicity features, we hypothesize that distinct, as well as similar, detrimental mechanisms in the respiratory system may occur, thus increasing the risk of respiratory depression from the co-ingestion of ketamine plus ethanol [44,64] (Figure 1D).

3.3. Urinary System

Urinary-system-related disorders have been correlated with the recreational use of ketamine [36,38,46,48]. The lower urinary tract has been the most affected region (early symptoms); interstitial cystitis presents the main evidence of ketamine misuse [48,53,72]. However, chronic ketamine use leads to more intense urinary symptoms and upper-tract involvement, such as obstructive uropathy and kidney damage [38,53]. Although both acute renal failure and hydronephrosis occur among ketamine abusers, such effects frequently are recovered during ketamine withdrawal [53,73]. The pathophysiology that underlies the ketamine-induced negative effects on the urinary tract is not well established; however, inflammatory abnormalities and the accumulation of precipitated ketamine metabolites in the pelvicalyceal systems may produce a direct toxicological effect on the urothelium, mainly at high plasmatic concentrations [53,74]. In a rat model, prolonged exposure to high dose of ketamine resulted in infiltrative inflammatory processes in the bladder and kidney, wherein the kidney seems to be more vulnerable at lower doses, and its abnormalities are more persistent [75]. In fact, controversial findings postulate that such an infiltrative process is induced by alterations in ion channel of the cells (i.e., Na+ channels, Ca2+-activated K+ channels, adenosine triphosphate-sensitive K+ channels, and Hyperpolarization-Activated Cyclic Nucleotide (HCN)-1 channels), but not by a ketamine deposit itself [76,77,78,79]. We hypothesize that an inflammatory process has been elicited by both mechanisms, the immune response and the disruption of ion channels’ homeostasis also resulted from the chemical presence of ketamine and its metabolites on the urinary system. In addition, in the long term, even in withdrawal after a long-lasting ketamine exposure, neurogenic damage associated with the loss of cholinergic neurons and neurotransmission reduction, as well as abnormalities on purinergic pathway, also underlie cystitis and bladder disruption [80,81].
Although there are conflicting theories related to the hazardous effects of ethanol on the urinary tract, the kidney seems to be the structure that is most vulnerable to alcohol’s detrimental effects. Regarding the lower urinary tract, robust evidence about the relationship between the deleterious effects of alcohol and ureters and the bladder is still scarce [82]. Thus, we focused on the upper urinary tract, specifically the kidney. In fact, epidemiological studies have linked heavy alcohol consumption as a modified risk factor for the development of renal damage [83,84]. Herein, experimental studies have investigated the pathophysiological features that underlie alcohol-induced kidney disorders [85]. Firstly, chronic alcohol exposure induces renal-structure changes, compatible with glomeruli atrophy, tubular necrosis, and alterations on renal tubules’ epithelia that can progress to renal fibrosis and glomerular sclerosis, primary induced by persistent hypertensive status and endothelial impairment [85,86,87,88]. In addition, renal function is mainly affected by hyperactivation of renin–angiotensin system, specifically mediated by the overexpression of angiotensin II and its receptor (angiotensin II type 1 receptor-AT1R), upregulation of the renal sympathetic pathway, and oxidative stress [85,89,90,91,92]. Besides the loss of renal homeostasis dysfunction, systemic and local elevation of blood pressure and renal failure occur in the long term, even in withdrawal [85,89,90,91,92].
Although ketamine abuse has been accompanied by alcohol intake among misusers, few clinical studies have underlined the toxicological effects that result from the association, focusing mainly on the urinary system [37,38]. In an experimental study, ketamine-plus-alcohol-treated animals developed atresia of glomeruli and necrotic cell in the kidney related to proteinuria, which infers renal dysfunction [61,62]. Such evidence suggests that ketamine–alcohol co-treatment augments the toxicological effects of both drugs per se, reducing the period of exposure to elicit nephrological impairment [62]. However, it is unclear the pathophysiology of such toxicological synergic mechanisms. We hypothesize that pro-inflammatory-pathway triggering of oxidative stress, ion channels’ homeostasis disruption, and elevation of renal blood pressure may underlie kidney damage in ketamine plus ethanol abuse (Figure 1E).
Finally, despite that the main molecular mechanisms investigated upon ketamine and alcohol occur in the CNS, the findings above highlight that these substances may affect other important organic systems; furthermore, they reinforce that peripherical alteration can elicit behavioral alterations. Both clinical and pre-clinical studies have shown that ketamine can display peripheral unbalance; however, clinical studies have key features that need to be pointed out. Works have failed to explore ketamine per se among users, since polydrug use is common in addiction. Moreover, it is evident that such a gap in the distinction of misused substances does not permit us to establish adequate drug-effect identification. Thus, experimental studies claim to identify such detrimental effects and have reported the occurrence of renal, hepatobiliary, cardiovascular, and respiratory damage, in addition to CNS injury discussed later. Clinical studies on ethanol’s harmful effects have been well documented, considering that alcohol consumption per se frequently occurs.
To facilitate the visualization of these changes, as well as to compilate relevant information about such relevant studies discussed until this point in the paper, we summarize, in Table 1, the effects of ketamine and/or ethanol on the liver, biliary, cardiovascular, respiratory, and urinary systems.

3.4. Central Nervous System

Ketamine CNS effects have been extensively described, primary by its psychotropic therapeutic use. Among recreational users, such a psychoactive drug at subanesthetic doses induces dissociative effects (also called “K-hole”), hallucinations, and altered status of consciousness, which favors the searching for this substance [93,94]. CNS consequences displayed by ketamine’s abuse range from cognitive-function disabilities to psychiatric disorders [93].
On the other side, as extensively discussed elsewhere, alcohol abusers undergo several behavioral alterations, thus reflecting its widespread toxicological effects on CNS, with long-lasting negative repercussions [95,96].

3.4.1. Behavioral Disorders

Psychosis

Curran and Morgan’s research group has extensively studied the hazardous effects of ketamine recreational use on the CNS [40,93,97,98,99,100,101,102,103,104,105,106,107]. Schizophrenic symptoms have been frequently reported in frequent users of ketamine [24,40]. The immediate effects after 30 min of ketamine consumption elicit markable levels of schizophrenia-like behavior in polydrug abusers, with magical ideation, perceptual distortion, and thought-disorder symptoms [24]. Of interest, in early withdrawal (i.e., 24 h or 3 days of abstinence), schizotypal symptomatology still persists, characterizing a residual repercussion [24,39,103]. These psychopathological features have appeared in frequent, as well as in infrequent, users, correlated to extensive use of ketamine [39]. Such a ketamine-induced schizophrenic profile has been related to the reduction of NMDA receptors, and this elicits the NMDA-dependent negative symptoms observed among schizophrenic individuals who are free of clinical treatment [108,109,110].
The exact mechanism that involves schizotypy symptoms has been investigated. NMDA receptors’ blockade on parvalbumin-expressing (PV) interneurons results in disinhibition of pyramidal cells and increment of γ-band oscillations, reflecting cortical hyperactivity that plays a pivotal role in schizophrenia manifestation, related to positive symptoms such as hallucinations, delusions, disorganized speech, and catatonic behavior observed during ketamine bioavailability [111,112,113]. In this context, ketamine administration may display hyperactivation of glutamatergic circuitry in the brain that is associated with CNS overstimulation, resulting in behavioral and cognitive impairments, which reflect schizophrenic features [114,115,116,117]. In the long term, mechanisms of prevention of glutamatergic excessive transmission and excitotoxicity are dull, since the excitatory amino acid transporters 1 and 2 (EAAT1/2) expressed in astrocytes, which are responsible for the reuptake of glutamate, undergo downregulation after NMDA blockers’ exposure, which supports the hyperlocomotion and other positive characteristics in withdrawal related to schizophrenic patients [118].
Although the hyperactivation of glutamatergic circuitry plays of a vital role in ketamine-induced psychomimetic manifestations, dopamine receptor D2-like hyperfunction strongly contributes to schizophrenic symptoms [119]. In fact, animal model studies reveal that NMDA-blockers elicit positive schizotypy by enhancing neurotransmitters’ release, not only glutamate and dopamine, but also serotonin and acetylcholine, in the prefrontal cortex [108,120,121,122]. In addition to having an NMDA-receptor-dependent mechanism, ketamine also directly activates dopamine D2-receptor, with equal affinity observed in the NMDA receptor, which strongly contributes to psychotomimetic effects [123]. Moreover, studies have evidenced ketamine’s weak binding affinity for 5-hydroxytryptamine (HT)2A receptors. In fact, 5-HT2A activation by ketamine occurs through the direct interaction with this receptor, as well as the inhibition of voltage-gated K+ channel (Kv) currents’ conductance modulation, potentiating Kv inhibition mediated by 5-HT2A receptor, facilitating both membrane depolarization and 5-HT2A receptor signaling [29,124]. Furthermore, the acetylcholine transmission system increases and triggers the release of additional serotonin in the prefrontal cortex, and this may result in schizophrenic features related to serotoninergic hyperactivation pathway, resulting in visual hallucinations [122,125].
Opioid receptors represent other weak pharmacological targets implicated in the pathophysiology of schizophrenia [126]. The hypothesis that the sigma-receptor is involved in the schizotypy displayed by ketamine use relies on controversial studies demonstrating that postmortem schizophrenic-brained individuals present reduced levels of sigma receptors in the temporal cortex, but not in the parietal area [127,128]. The hyperactivation of sigma receptors (specially sigma-1 receptor) by phencyclidine (PCP)-like drugs may contribute to schizophrenic-like symptoms observed in ketamine abusers [126]. In addition, the kappa opioid receptor that displays psychomimetic properties presents ketamine binding affinity, which also may contribute to schizophrenic symptoms [129,130]. Finally, we hypothesize that promiscuous ketamine targets, primarily at NMDA, D2-type, and 5-HT2A receptors, as well as other weaker sites, may synergically potentiate psychotic effects in ketamine abusers (Figure 2).
Although alcohol psychotic-like symptoms are difficult to diagnose because of confounding elements (i.e., alcohol withdrawal delirium, which presents a short course of illness), it is well documented that alcohol addiction is an important aetiologic factor for psychosis, especially the auditory hallucinatory symptoms associated with delusions, as described since 1847 [131,132]. This condition is called Alcohol-Induced Psychotic Disorder by the American Psychiatric Association (DSM V-TR, 2000) [133]; the discussion relied on whether alcohol acts as a trigger factor for schizophrenia in a latent form [134]. This axiom has been rejected after differential diagnosis proposed by specialist authors [135]. Anxiety, auditory hallucinations, and delusions of persecution are the features related to alcohol hallucinosis syndrome, which can result in paranoid psychosis [132]. The pathophysiology of Alcohol-Induced Psychotic Disorder presents several gaps. Few image studies show the frontal, thalamic, basal ganglia, and cerebellar regions’ reduced activity during psychotic symptoms in withdrawal [136,137].
Alcohol-induced hallucinations share several of the neurobiological events elicited by psychotic-like ketamine-induced symptoms. During both ethanol and ketamine blood concentrations, dopaminergic circuitry hyperactivation, serotonin brain reduction, and reduced inhibitory transmitters (i.e., GABA and glycine) contribute to hallucinations displayed by chronic alcohol intake [138,139,140] (Figure 2).
Furthermore, ethanol and ketamine withdrawal display hyperactivation of the brain excitatory pathway after a prolonged blockade of excitatory receptors (especially NMDA receptors) [138] (Figure 3). We suggest that these overlapping molecular toxicological mechanisms may synergically contribute to psychotic-like effects among users of ketamine in associated with alcohol. Actually, a preclinical study demonstrated that co-exposure of ketamine plus alcohol for 14 consecutive days increased the release of dopamine and glutamate in the cortex and hippocampus in relation to hyperlocomotion, which characterizes the schizotypy profile during both drugs’ bioavailability [141]. Moreover, alcohol-potentiated neurotoxicity is induced by ketamine administration through mitochondrial dysfunction; brain-derived neurotrophic factor (BDNF) signaling impairment; and inhibition of cyclic AMP-responsive element binding protein (CREB) pathways’ signaling factors, i.e., serine/threonine kinase (Akt), calmodulin-dependent kinase IV (CaMKIV), and protein kinase A (PKA), along with reflexes on apoptosis, synaptic plasticity, and neuronal growth [141] (Figure 3). Of note, the influence of co-exposure exhibits distinct responses related to dopamine release, as well as BDNF expression, according to the specific brain tissue and the dose of both toxicants [142].

Depression

Although ketamine’s antidepressant effects have been explored, a longitudinal study among non-medical users has reported that mood depression disorder was prevalent even after a prolonged absence of the drug [40]. In fact, in a relevant controlled clinical study conducted among ketamine non-medical users, Morgan and colleagues found that frequent users (i.e., ketamine self-administration at least four times/week) presented depressive symptoms in withdrawal, at a mild-to-moderate scale, in a dose-dependent manner [39]. In a subsequent study in withdrawal, the authors showed that depression disorder was present in both frequent and abstinent ketamine users, reflecting changes in longitudinal emotional effects, even in the absence of depressive features previously detected [39,40]. Such relevant findings highlight the risk of the development of additional long-lasting psychological disorders across the time after ketamine misuse, correlated to a pattern of self-administration, which is, curiously, since ketamine has been claimed to have prolonged antidepressant activity in depressive patients [105]. It is noteworthy to mention that, even at therapeutical doses, ketamine elicits psychotic symptoms in healthy volunteers, thus suggesting its potential to modulate mental status in a way that leads to disordered thinking and behavior in healthy individuals [108]. Nonetheless, under abuse conditions, the frequency of consumption, dose, route of administration, and co-administration with others psychoactive substances are relevant factors that affect the negative impairments observed [108].
The pathophysiological mechanism that underlies depression symptomatology linked to the recreational use of ketamine appears to be a result of combinate mechanisms. Since ketamine acts as an antagonist of NMDA receptors, a compensatory increase of glutamate activity may occur, as usually observed in NMDA-blocker drugs, including ethanol, resulting in excitotoxicity in withdrawal period [143,144,145] (Figure 3). In turn, excitotoxicity underlies neurodegenerative processes, marked by microglial activation and astrocytic hypertrophy, which culminate in increased levels of pro-inflammatory cytokines, induction of cyclooxygenase 2, upregulation of neuronal nitric oxide synthase, and oxidative stress in animal models [146]. All of these processes have been claimed as molecular mechanisms that elicit depression [147,148,149,150] (Figure 3). Actually, our group showed that ketamine withdrawal elicits depressive-like behavior associated to hippocampal oxidative damage, thus suggesting that oxidative stress plays a role in the neurobehavioral impairment induced by the ketamine subanesthetic paradigm in animal models [25].
On the other hand, ketamine also induces the activation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, triggering the expression of their subunit GluA1, as well as stimulating the mammalian target of rapamycin (mTOR), which consists of mechanisms involved in synaptic plasticity and synaptogenesis [108]. Bioavailable ketamine inhibits NMDA receptors and upregulates AMPA receptors on medial prefrontal cortex pyramidal cells, resulting in the activation of projections to dorsal raphe nucleus and serotonin release [108] (Figure 2). Thus, it is reasonable to infer that ketamine bioavailability and withdrawal affect neuronal plasticity and decrease serotonin signaling, which directly affects emotional homeostasis (Figure 2 and Figure 3).
The literature has proposed that ketamine inhibits the neuronal reuptake of norepinephrine, dopamine, and serotonin [65]. The increased levels of dopamine and noradrenaline observed after ketamine administration suggest the existence of stimulatory projections to the ventral tegmental area and locus coeruleus [108], as illustrated in Figure 2. Particularly, an increase of dopamine activity in the Nucleus accumbens is involved in addiction mechanisms of several drugs’ abuse, according to the cerebral reward system theory, which proposes that the absence of these psychoactive substances consequently induces depressive symptoms [151,152] (Figure 3). In fact, ketamine per se increases dopamine levels related to rewarding behavior in a place-preference task, which characterizes the potential dependence effect of this drug [19] (Figure 2).
In addition, depression has been extensively documented during withdrawal among alcohol abusers in clinical studies, as well as in animal models. It has been proposed that the overstimulation of NMDA receptors occurs in major depression [108]. Moreover, studies report compensatory effects of glutamate activity during ethanol abstinence, which induces neuronal excitotoxicity through NMDA receptor hyperactivation [95,143,144,145,153,154,155] (Figure 3). As mentioned before, excitotoxicity results in neuronal degeneration and neuroinflammation, in which such disturbances have been widely reported in depression pathology [108,146,148,149]. Indeed, depression symptomatology has been associated with cytokine and chemokine expression levels’ upregulation in the saliva and plasma of individuals that consumed ethanol, as well as dysbiosis of salivary microbiota [156]. Furthermore, a degenerative process provoked by excitotoxicity is accompanied by mitochondrial damage, release of superoxide radicals, lipid peroxidation, and other mechanisms linked to oxidative stress [147,150]. In experimental studies, our group found depressive-like behavior during withdrawal in rats submitted to binge-drinking protocol during adolescence, related to oxidative stress [21,157] (Figure 3). Robust evidence has proposed that depression pathophysiology is associated with reduction of neuronal viability, neuroinflammation, and oxidative stress, and may explain the occurrence of depressive profile among chronic ethanol users [95,146,147,148,149,150]. Finally, ethanol also alters the activity of several monoamines related to mood (monoamine theory), thus contributing to depressive outcomes [158,159,160,161] (Figure 3).
The isolated effects of ethanol and ketamine have direct or indirect potential to trigger depressive symptoms. However, few studies have explored the concomitant consumption consequences of these toxicants. Apparently, under the bioavailability of both of these drugs, reinforcement of compensatory effects in glutamatergic activity occurs, resulting in neuronal apoptosis stimuli and downregulation of important pathways of neuronal plasticity on cortex and hippocampus in animals [141]. As commented previously, hyperactivation of NMDA–glutamatergic signaling provokes excitotoxicity, which leads to apoptosis, neurodegeneration, neuroinflammation, and oxidative stress, including in brain areas related to emotional behavior, favoring depressive status in withdrawal [162,163] (Figure 3). In addition, disturbances in monoamines’ activities and synaptic plasticity may alter neurotransmission homeostasis, thus eliciting depression symptoms. In a 30-days ketamine-plus-ethanol challenge withdrawal, animals exhibited rewarding effects in a drug-abuse paradigm. [19]. This altered behavior was associated with dopamine levels being increased in then striatum, the upregulation of four dopamine metabolism genes mRNA levels, and BDNF overexpression on the cortex–striatum circuitry [19]. This augment of dopamine levels elicited by ketamine-plus-ethanol exposure also was reported in the hippocampus [141]. These findings are aligned with the monoamine theory of depression [164], and they also support the predictive potential rewarding behavior of these psychoactive drugs in co-misuse, overstimulating the rewarding pathway, and consequently developing depressive symptoms in long-lasting withdrawal (Figure 3).
In addition, chronic ketamine-plus-ethanol co-administration for 21 days in adolescent rats resulted in depressive-like behavior, an increase of apoptotic cells, and the upregulation of the expression of pro-apoptotic proteins caspase-3 and Bax in prefrontal cortex during withdrawal [164]. In Li et al.’s study, these negative effects were intensified by co-intoxication [164]. These data reflect the pro-apoptotic process as another neurotoxicological mechanism elicited by co-abuse in depression pathophysiology (Figure 3).

Anxiety

Anxiety is a complex emotional disturbance, and its pathophysiology involves central and autonomic responses; it is a common manifestation in CNS disorders. Among ketamine users, anxiety, accompanied by shaking and sweating, is a symptom that is manifested during withdrawal [93]. In a clinical study with ketamine users during a hospital detoxification treatment, ketamine chronic users exhibited moderate anxiety [165]. In an additional longitudinal study, ketamine users’ individuals presented high scores of anxiety profile in anxiety-test measurement [166]. In animal models, controversial data were found. Acute administration in the first hours of consumption elicits a rapid, but not sustained, anxiolytic response, while long-lasting evaluations and chronic use induced anxiety in withdrawal [167].
Our group showed that ketamine three-consecutive-administration protocol withdrawal elicits an anxiety-like profile in adolescent rats that is associated with oxidative damage in the hippocampus [25]. Oxidative stress and neuroinflammation are common components of the pathology of several CNS disorders. Considering the possible neuroadaptations due to the multiple systems affected by ketamine, especially the NMDA receptor blockade, excitotoxicity might occur (Figure 2). Damage in essential brain areas related to fear and alertness control (i.e., prefrontal cortex) under both drugs’ effects or withdrawal in animals was observed, for which the induction of apoptosis and reduction of neuronal viability may disrupt physiological mechanisms of anxiety inhibition [141,168] (Figure 2 and Figure 3).
Glutamatergic hyperfunction have been proposed to evoke anxiety-related symptoms in ethanol users during an abstinence period [143,154] (Figure 3). In addition, sympathetic and cortisol overactivity represent the consequences postulated by hypothalamus–pituitary–adrenal (HPA) axis activation theory, as elements of the pathophysiology of stress and anxiety [169]. In the ketamine literature, we did not find studies that investigated such pathway; however, some studies have demonstrated that the HPA axis is affected by ethanol exposure [96,170,171,172]. Finally, dysregulations in monoamine activities also can be speculated, since drugs that equilibrate the release and functions of these neurotransmitters are able to control symptoms manifested by these anxious patients [65,108,161]. Previously, we discussed the effects of both drugs on monoamines homeostasis, especially during withdrawal; however, it is necessary clarify if and how this imbalance occurs and contributes to anxiety manifestations. For example, ketamine induces an increase in monoamine release, and the opposite effect was expected in withdrawal [65] (Figure 2 and Figure 3). On the other hand, it has been proposed that serotonin transmission increases after acute alcohol administration and decreases during alcohol abstinence, and this might contribute to anxiogenic symptoms in withdrawal [161] (Figure 3).
Although studies have failed to prove the anxiogenic effect under ketamine plus ethanol bioavailability (1 h post-administration) or during withdrawal (24 h post-administration) [168,173], peculiar factors such as the dose utilized, treatment duration, and time of evaluation after intoxication may influence the results obtained; thus, additional studies are necessary to clarify the comprehension of potential toxicological effects of this co-abuse in relation to anxiety. We speculate that the convergence of toxicological mechanisms shared by ketamine and ethanol on anxiety neurobiology presents potential synergistic effects, which theoretically can intensify the pathophysiological mechanisms of anxiety, resulting in an amplification of anxiety symptoms. For example, synergic activities elicited by ketamine and ethanol resulted in neurotoxicity induced by glutamatergic pathway upregulation, as well as neuronal loss on cortical areas [83,141,144,145]. In fact, it remains unclear how these synergistic effects on the monoaminergic system contribute to anxiety.

Cognitive Disorders

Scarce studies have focused on ketamine and its consequences on cognition. Experimental studies have found that a single neonatal exposure to the therapeutic use of ketamine leads to short-term reduction in hippocampal cellular viability, as well as long-term alterations in hippocampal glutamate transport and short-term recognition memory prejudice in withdrawal [174]. Clinically, ketamine’s therapeutic use as an anesthetic agent in a pediatric patient in a pediatric intensive care unit displayed long-lasting disabilities in cognition [175]. In this sense, early exposure, even under therapeutic use, might provoke disruptions on cognitive domains, especially related to glutamatergic pathway inhibition.
In regard to non-therapeutic use, Morgan and colleagues have reported semantic and episodic memory compromising among ketamine misusers’ withdrawal, evaluated 3 days following drug use and in a follow-up after 3 years [105]. These authors observed that, even after the reduction of ketamine consumption over 3 years following the first assessment, impairments in attention and episodic memory still persist [105].
As mentioned previously, ketamine blocks NMDA receptors, which are responsible for crucial steps of memory formation, such as the long-term potentiation (LTP) process. LTP is classically described as being related to hippocampal neurons of the CA1 region of the dentate gyrus in a process mediated by glutamate, which activates the NMDA receptor that, in turn, induces extracellular signal regulated protein kinase (ERK) activation, followed by cAMP-responsive element-binding protein (CREB) phosphorylation, which persists for hours after LTP induction [176,177] (Figure 2). CREB promotes memory formation through the upregulation of neuronal excitability, memory trace cells activity, and successive events associated to memory storage and consolidation [178,179,180]. In addition, CREB phosphorylation induces the expression of BDNF, an important neurotrophin that plays a crucial role in synaptic activity transformation into long-term synaptic memories and general synaptic plasticity through interaction with tropomyosin related kinase (Trk) receptors, especially TrkB. BDNF/TrKB signaling activates downstream cascades, such as phosphatidylinositol 3-kinase (PI3-K)/Akt pathways, resulting in adequate cognitive process [178,179,180]. Thus, a blockade of NMDA receptors may result in cognitive deficits during ketamine bioavailability, as well as during withdrawal (Figure 2 and Figure 3). Indeed, as mentioned above, ketamine alters dopaminergic responses. There is evidence that, during abstinence, chronic ketamine consumers exhibited D1-receptor regional selective upregulation availability in dorsolateral prefrontal cortex, a phenomenon caused by chronic dopamine depletion in animal studies [181]. This fact indicates that the repeated use of ketamine impairs prefrontal dopaminergic signaling, which is a crucial step in working memory and executive function [181], as shown in Figure 2.
In turn, alcohol presents the NMDA receptor blockade as a primordial mechanism of action on the CNS; however, this toxicant can disrupt the homeostasis of several systems, and this can lead to cognitive impairment [96] (Figure 2). An extensive amount of works in the literature have shown the cognitive consequences elicited by ethanol consumption, affecting most processes related to cognition, even as a residual deleterious effect in long-term abstinence [182] (Figure 3).
Thus, it is reasonable to infer that the co-intoxication by ketamine plus ethanol may display profound consequences in cognitive domains and/or presents a risk augmented of mnemonic damage that is even higher than both substances isolated. Recently, it was demonstrated that ethanol aggravates ketamine-induced neurotoxicity through the downregulation of Akt, CREB, protein kinase A (PKA), and calmodulin-dependent kinase IV (CaMK-IV) on the cortex and hippocampus of adolescent rats under both substance effects [141] (Figure 2). These proteins are crucial for cell survival, and the repression of such signaling impairs neuronal viability and plasticity, thus affecting cognitive function [182,183,184]. Thus, a long-term negative impact on CREB and its downstream cascades can interfere directly with synapsis function and memory storage, even in withdrawal (Figure 3).
Finally, neuroadaptations resulting from ketamine-plus-ethanol withdrawal might be considered. We highlight the impact of events such as glutamatergic excitotoxicity and how subjacent apoptosis, neuroinflammation, oxidative stress, etc., might disrupt cognitive homeostasis (Figure 2). Findings related to ketamine and ethanol inducing similar damages support this theory [83,141,144,145,161,174,185]. It is noteworthy that these events have been extensively reported in the literature as components of pathophysiology of important cognitive disorders, such as Alzheimer’s disease [186,187,188] (Figure 2). Table 2 presents a compilation of studies related to the effects of ketamine or/and ethanol on the CNS in which polydrug consumption information was included.

4. Potential Pharmacological Interactions between Ketamine and Ethanol Use

The context of interaction between ethanol and ketamine is considerably complex. In fact, ethanol is the most consumed psychoactive drug worldwide, and it is culturally and legally accepted in most countries, with it mainly being linked to recreational contexts [96]. The broad variation in patterns of dose, frequency, and intermittence of consumption increases the complexity of analysis, since such patterns (i.e., acute effects vs. chronic consumption) induce opposite outcomes [6,7,8,9]. Ketamine, on the other hand, presents its use primarily linked to a medical context, as a sedative analgesic and general anesthetic, and more recently being proposed for affective disorders treatment [189,190]. However, ketamine has become popular as a recreational drug, used acutely or in a binge pattern; orally, inhaled, or injected; and alone or associated with alcoholic beverages or stimulants [191]. Each of these paradigms must be examined to identify potential synergistic points and risks related to this combination [11,12,13].
Pharmacokinetically, interactions between ethanol plus ketamine consumption are reasonable through competition for the CYP3A4 metabolic enzyme [192,193]. Although such an enzyme is not the primary route for alcohol metabolism, the consumption of a large amount of ethanol, as occurs in binge drinking pattern, elicits a metabolic enzymes saturation phenomenon, which may reduce ketamine metabolism, increasing ketamine bioavailability. On the other hand, ethanol chronic consumption induces CYP3A4 expression, increasing ketamine biotransformation and consequently reducing bioavailability. Therefore, investigations are necessary to elucidate the consequences of these interactions and their clinical relevance.
As highlighted in this review, systemic toxicity of both drugs is another point that deserves attention. Interestingly, both ethanol and ketamine present coincidental or synergistic deleterious effects on cardiorespiratory, hepatic, and urinary tracts [46,51,55,56,57,58,60,62,64,67,71,82,83]. Some of these damages are linked to concurrent pharmacological mechanisms on glutamatergic, noradrenergic, and cholinergic pathways’ modulation, resulting in biliary motility impairment, tachycardia, respiratory disability, and intraglomerular pressure. The ability to modulate ion channels, mitochondrial function, and oxidative metabolism interference is also common to both drugs, frequently triggering cell dysfunctions [194]. Ionic dysregulation is correlated with pulmonary edema, cardiac arrhythmias, and urinary dysfunction [195]. Mitochondrial dysfunction is associated with pro-oxidant effects that are related to inflammation, cell injury, apoptosis, and fibrosis in different systems [196,197]. Such harmful mechanisms shared by ethanol and ketamine use highlight the potential risk of this association that requires further investigation.
In CNS, the harmful potential effects of both of these drugs are mainly related to the concomitant ability to modulate glutamatergic and GABAergic pathways [198]. Different manifestations and symptoms may emerge, depending on the pattern of use (under effect of the drugs) or abstinence symptoms’ consequences. Considering that binge consumption consists of the main pattern among misusers, both ketamine and ethanol display a reduction of glutamatergic activity on NMDA receptors [198]. Additionally, ethanol potentiates GABAa receptors’ activity. Therefore, interference in formation of LTP processes, as well as reduced production of BDNF and Bcl-2, has been observed, impairing neuroplasticity and memory consolidation [199]. Accessory mechanisms such as the modulation of dopaminergic, serotonergic, cholinergic, and pathways, among other things, also may be shared by the drugs per se [19,65,96,108,158,159,160,181]. Considering that the isolated use of these substances, according to dose, induces distinct levels of cognition and perception disturbance, also identifying molecular synergistic potential, theoretically, the co-use may aggravate outcomes; this deserves further studies to be properly proved.
In an abstinence context, ketamine and ethanol also share deleterious mechanisms, involving overspread glutamatergic excitotoxicity, resulting in oxidative stress, mitochondrial dysfunction, neuroinflammation, and cell death induction [200,201]. These hazardous processes have been associated with genesis and poor outcomes of several CNS diseases, such as depression and anxiety [108,146,147,148,150]. Once again, the severity of manifestations might be influenced by dose, frequency, and intermittence of use, but the correlation between isolated use of ketamine or ethanol vs. anxiogenic or depressive-like behavior manifestation has already been demonstrated [25,202]. Based on all convergent targets divided by ethanol and ketamine use, which we have highlighted above in the present review, the probable risk of potentiation of CNS disorders in co-use circumstances is imminent and deserves further research.

5. Conclusions

The abuse of psychoactive substances has increased worldwide, catalyzed by economic and social factors, in addition to global calamities that affect humanity. In contrast to alcohol, which consists of the most consumed psychoactive drug, ketamine misuse has been augmented, attributed to its psychedelic activities. However, the co-ingestion of these drugs occurs among individuals who ignore the toxicological potentials of each drug and the risks of their combined use. Consequences of toxicological mechanisms shared by ketamine and ethanol might intensify several body systems’ damages, which can culminate in fatal outcomes. In the gastrointestinal system, concomitant abuse displays cirrhosis and hepatobiliary symptoms. Cardiorespiratory negative consequences, such as cardiotoxicity and respiratory depression, are the main evidence of toxicological hazardous effects. In the urinary system, renal dysfunction represents a critical outcome resulted from ketamine plus ethanol consume. Although the literature focused on the consequences of ketamine plus ethanol abuse is scarce, for the CNS, we can find extensive works in the literature. Similar mechanisms of action shared by ketamine and ethanol may potentiate the toxicological disturbance, resulting in schizotypy symptoms, depression, anxiety, and cognitive impairment. Additional studies are urgently needed to minimize the gap in the literature related to this co-abuse, as well as to explore future strategies to reduce the toxicological consequences that are caused by ketamine plus ethanol misuse.
As highlighted above, there are a variety of targets with potential for harmful interaction, compromising hepatobiliary, respiratory, cardiac, and urinary function, and thus placing users’ lives at risk. Similarly, several mechanisms related to changes in consciousness, cognition, and emotionality are shared by ketamine and ethanol, related to use and consequent repercussions in withdrawal. The consequences of such co-abuse deserves to be properly investigated in order to elucidate the toxicological mechanisms and clinical outcomes, clarify them to the community, and propose the development of new therapeutic strategies.

Author Contributions

Conceptualization, C.d.S.F.M.; methodology, C.d.S.F.M.; software, E.A.F.-J. and B.C.d.C.; resources, C.d.S.F.M., N.H.C.K., S.V.F. and C.C.M.d.S.; writing—original draft preparation, C.d.S.F.M., N.H.C.K. and S.V.F.; writing—review and editing, C.d.S.F.M., D.A.L., K.M.M.-F., L.M.P.F., S.d.C.C. and V.M.M.F.; visualization, C.d.S.F.M., N.H.C.K., S.V.F. and C.d.S.F.M.; figures design, E.A.F.-J. and B.C.d.C.; supervision, C.d.S.F.M.; project administration, C.d.S.F.M.; funding acquisition, C.d.S.F.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by a Research Productivity Grant awarded to Cristiane do Socorro Ferraz Maia by the Conselho Nacional de Desenvolvimento Científico e Tecnológico-CNPq/Brazil CNPq, grant number 311335/2019-5; and by the Research Pro-Rectory of the Federal University of Pará (PROPESP, UFPA, Brazil), which provided the article publication fee.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Olsen, Y. What Is Addiction? History, Terminology, and Core Concepts. Med. Clin. 2022, 106, 1–12. [Google Scholar] [CrossRef] [PubMed]
  2. Bombana, H.S.; Bogstrand, S.T.; Gjerde, H.; Jamt, R.E.G.; de Carvalho, H.B.; Andreuccetti, G.; de Oliveira Bernini, C.; Muñoz, D.R.; Leyton, V.; Greve, J.M.D. Use of alcohol and illicit drugs by trauma patients in Sao Paulo, Brazil. Injury 2022, 53, 30–36. [Google Scholar] [CrossRef] [PubMed]
  3. Wagner, V.; Flores-Aranda, J.; Villela Guilhon, A.C.; Knight, S.; Bertrand, K. How do Past, Present and Future Weigh into Trajectories of Precarity? The Time Perspectives of Young Psychoactive Substance Users Living in Situations of Social Precarity in Montreal. Qual. Health Res. 2022, 32, 195–209. [Google Scholar] [CrossRef] [PubMed]
  4. Dos Santos, A.R.; Perrelli, J.G.A.; Silva, T.T.D.M.; Lopes, M.V.D.O.; Frazão, I.D.S. Instruments related to drug use in adolescents: An integrated review. Texto Contexto Enfermagem. 2018, 27, e0370017. [Google Scholar]
  5. Silva, J.L.; Silva, R.L.; Silva, A.L.; Silva, G.C.B.e.; Cortez, A.C.L. Uso de Substâncias Psicoativas “Drogas”: Uma Revisão De Literatura/Psychoactive Substance Use “Drugs”: A Review. Northeast. Braz. Health J. 2013, 1, 2–8. [Google Scholar]
  6. Cohen, S.M.; Alexander, R.S.; Holt, S.R. The Spectrum of Alcohol Use: Epidemiology, Diagnosis, and Treatment. Med. Clin. N. Am. 2022, 106, 43–60. [Google Scholar] [CrossRef] [PubMed]
  7. World Health Organization. Alcohol; World Health Organization: Geneva, Switzerland, 2018. [Google Scholar]
  8. Manzo-Avalos, S.; Saavedra-Molina, A. Cellular and mitochondrial effects of alcohol consumption. Int. J. Environ. Res. Public Health 2010, 7, 4281–4304. [Google Scholar] [CrossRef] [Green Version]
  9. Rehm, J.; Mathers, C.; Popova, S.; Thavorncharoensap, M.; Teerawattananon, Y.; Patra, J. Global burden of disease and injury and economic cost attributable to alcohol use and alcohol-use disorders. Lancet 2009, 373, 2223–2233. [Google Scholar] [CrossRef]
  10. Marco, C.A.; Sich, M.; Ganz, E.; Clark, A.N.J.; Graham, M. Penetrating trauma: Relationships to recreational drug and alcohol use. Am. J. Emerg. Med. 2022, 52, 8–12. [Google Scholar] [CrossRef]
  11. Vwaire Orhurhu, A.J.; Vashisht, R.; Claus, L.E.; Cohen Afiliações, S.P. Toxicidade da Cetamina Atividade de Educação Continuada. Available online: https://www.ncbi.nlm.nih.gov/books/NBK541087/?report=printable (accessed on 26 March 2022).
  12. Lee, T.S.H.; Liu, Y.H.; Huang, Y.J.; Tang, W.K.; Wang, Y.; Hu, S.; Lin, C.P.; Li, C.S.R.; Hung, C.C. Clinical and behavior characteristics of individuals who used ketamine. Sci. Rep. 2022, 12, 1–8. [Google Scholar] [CrossRef]
  13. UNODC—United Nations Office on Drug and Crime. World Drug Report; United Nations Office on Drug and Crime: Vienna, Asutria, 2015. [Google Scholar]
  14. Peoples, R.W.; White, G.; Lovinger, D.M.; Weight, F.F. Ethanol inhibition of N-methyl-D-aspartate-activated current in mouse hippocampal neurones: Whole-cell patch-clamp analysis. Br. J. Pharmacol. 1997, 122, 1035–1042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Kaczmarek, B.; Kowalski, K.; Bogudzińska, B.; Piotrowski, P. Ketamine as A Novel Drug for Depression Treatment. Psychiatr. Danubina 2021, 33, 468–474. [Google Scholar] [CrossRef] [PubMed]
  16. Roberto, M.; Kirson, D.; Khom, S. The role of the central amygdala in alcohol dependence. Cold Spring Harb. Perspect. Med. 2021, 11, 1–23. [Google Scholar] [CrossRef] [Green Version]
  17. Honey, C.R.; Miljkovic, Z.; Macdonald, J.F. Ketamine and Phencyclidine Cause a Voltage-Dependent Block of Responses To L-Aspartic Acid. Neurosci. Lett. 1985, 61, 135–139. [Google Scholar] [CrossRef]
  18. Macdonald, J.F.; Miljkovic, Z.; Pennefather, P. Use-Dependent Block of Excitatory Amino Acid Currents in Cultured Neurons by Ketamine. J. Neurophysiol. 1987, 58, 251–266. Available online: www.physiology.org/journal/jn (accessed on 28 March 2022). [CrossRef] [PubMed]
  19. Liu, Q.; Xu, T.Y.; Zhang, Z.B.; Leung, C.K.; You, D.Y.; Wang, S.W.; Yi, S.; Jing, Q.; Xie, R.F.; Li, H.J.; et al. Effects of co-administration of ketamine and ethanol on the dopamine system via the cortex-striatum circuitry. Life Sci. 2017, 179, 1–8. [Google Scholar] [CrossRef] [PubMed]
  20. Fernandes, L.M.P.; Cartágenes, S.C.; Barros, M.A.; Carvalheiro, T.C.V.S.; Castro, N.C.F.; Schamne, M.G.; Prediger, R.D.; Monteiro, M.C.; Fontes-Júnior, E.A.; Cunha, R.A.; et al. Repeated cycles of binge-like ethanol exposure induce immediate and delayed neurobehavioral changes and hippocampal dysfunction in adolescent female rats. Behav. Brain Res. 2018, 350, 99–108. [Google Scholar] [CrossRef]
  21. Fernandes, L.M.P.; Lopes, K.S.; Santana, L.N.S.; Fontes-Júnior, E.A.; Ribeiro, C.H.M.A.; Silva, M.C.F.; de Oliveira Paraense, R.S.; Crespo-López, M.E.; Gomes, A.R.Q.; Lima, R.R.; et al. Repeated cycles of binge-like ethanol intake in adolescent female rats induce motor function impairment and oxidative damage in motor cortex and liver, but not in blood. Oxid. Med. Cell. Longev. 2018, 2018, 1–15. [Google Scholar] [CrossRef] [Green Version]
  22. Schmidt, K.J.; Doshi, M.R.; Holzhausen, J.M.; Natavio, A.; Cadiz, M.; Winegardner, J.E. Treatment of Severe Alcohol Withdrawal. Ann. Pharmacother. 2016, 50, 389–401. [Google Scholar] [CrossRef]
  23. Siomek-Gorecka, A.; Dlugosz, A.; Czarnecki, D. The molecular basis of alcohol use disorder (Aud). genetics, epigenetics, and nutrition in aud: An amazing triangle. Int. J. Mol. Sci. 2021, 22, 4262. [Google Scholar] [CrossRef]
  24. Curran, H.V.; Morgan, C. Cognitive, dissociative and psychotogenic effects of ketamine in recreational users on the night of drug use and 3 days later. Addiction 2000, 95, 575–590. [Google Scholar] [CrossRef] [PubMed]
  25. De Carvalho Cartágenes, S.; Fernandes, L.M.P.; Carvalheiro, T.C.V.S.; de Sousa, T.M.; Gomes, A.R.Q.; Monteiro, M.C.; de Oliveira Paraense, R.S.; Crespo-López, M.E.; Lima, R.R.; Fontes-Júnior, E.A.; et al. “Special K” drug on adolescent rats: Oxidative damage and neurobehavioral impairments. Oxid. Med. Cell. Longev. 2019, 2019, 5452727. [Google Scholar] [CrossRef] [PubMed]
  26. Hase, A.; Erdmann, M.; Limbach Hasler, G. Analysis of recreational psychedelic substance use experiences classified by substance. Psychopharmacology 2022, 239, 643–659. [Google Scholar] [CrossRef] [PubMed]
  27. Pan American Health Organization—PAHO. Alcoohl, Key Facts. Available online: https://www.paho.org/en/topics/alcohol (accessed on 1 February 2022).
  28. National Institute on Alcohol Abuse e Alcoholism (NIAAA); Department of Health and Human Services; National Institutes of Health NIAAA Newsletter; National Institute on Alcohol Abuse and Alcoholism. Featuring Information from the National Institute on Alcohol Abuse and Alcoholism; Office of Research Translation and Communications, NIAAA, NIH, DHHS: Bethesda, MD, EUA, 2004.
  29. Lin, H.; Kim, J.G.; Park, S.W.; Noh, H.J.; Kim, J.M.; Yoon, C.Y.; Woo, N.S.; Kim, B.; Cho, S., II; Choi, B.H.; et al. Enhancement of 5-HT2A receptor function and blockade of Kv1.5 by MK801 and ketamine: Implications for PCP derivative-induced disease models. Exp. Mol. Med. 2018, 50, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Molero, P.; Ramos-Quiroga, J.A.; Martin-Santos, R.; Calvo-Sánchez, E.; Gutiérrez-Rojas, L.; Meana, J.J. Antidepressant Efficacy and Tolerability of Ketamine and Esketamine: A Critical Review. CNS Drugs 2018, 32, 411–420. [Google Scholar] [CrossRef] [PubMed]
  31. Van Amsterdam, J.; van den Brink, W. Harm related to recreational ketamine use and its relevance for the clinical use of ketamine. A systematic review and comparison study. Expert Opin. Drug Saf. 2022, 21, 83–94. [Google Scholar] [CrossRef]
  32. Siegel, R.K. Phencyclidine and ketamine intoxication: A study of four populations of recreational users. Public Health Serv. Alcohol. 1978, 21, 119–147. [Google Scholar]
  33. Sassano-Higgins, S.; Baron, D.; Juarez, G.; Esmaili, N.; Gold, M. A review of ketamine abuse and diversion. Depress. Anxiety 2016, 33, 718–727. [Google Scholar] [CrossRef]
  34. Larabi, I.A.; Fabresse, N.; Etting, I.; Nadour, L.; Pfau, G.; Raphalen, J.H.; Philippe, P.; Edel, Y.; Alvarez, J.C. Prevalence of New Psychoactive Substances (NPS) and conventional drugs of abuse (DOA) in high risk populations from Paris (France) and its suburbs: A cross sectional study by hair testing (2012–2017). Drug Alcohol Depend. 2019, 204, 107508. [Google Scholar] [CrossRef]
  35. Johnston, L.D.; O’malley, P.M.; Miech, R.A.; Bachman, J.G.; Schulenberg, J.E. Sponsored by The National Institute on Drug Abuse at the National Institutes of Health. Monitoring the Future National Survey Results on Drug Use; Institute for Social Research, University of Michigan: Ann Arbor, MI, USA, 1975. [Google Scholar]
  36. Pavarin, R.M.; Marani, S.; Turino, E. Ketamine abusers referring to emergency departments in northern Italy: A cross-sectional study. Ann. Ist. Super. Sanita 2019, 55, 338–344. [Google Scholar]
  37. Wood, D.M.; Nicolaou, M.; Dargan, P.I. Epidemiology of Recreational Drug Toxicity in a Nightclub Environment. Subst. Use Misuse 2009, 44, 1495–1502. [Google Scholar] [CrossRef] [PubMed]
  38. Winstock, A.R.; Mitcheson, L.; Gillatt, D.A.; Cottrell, A.M. The prevalence and natural history of urinary symptoms among recreational ketamine users. BJU Int. 2012, 110, 1762–1766. [Google Scholar] [CrossRef] [PubMed]
  39. Morgan, C.J.A.; Muetzelfeldt, L.; Curran, H.V. Ketamine use, cognition and psychological wellbeing: A comparison of frequent, infrequent and ex-users with polydrug and non-using controls. Addiction 2009, 104, 77–87. [Google Scholar] [CrossRef] [PubMed]
  40. Morgan, C.J.A.; Muetzelfeldt, L.; Curran, H.V. Consequences of chronic ketamine self-administration upon neurocognitive function and psychological wellbeing: A 1-year longitudinal study. Addiction 2010, 105, 121–133. [Google Scholar] [CrossRef] [PubMed]
  41. Galea, S.; Coffin, P.O.; Ahern, J.; Leon, A.C.; Vlahov, D.; Tardiff, K. Opiates, cocaine and alcohol combinations in accidental drug overdose deaths in New York City 1990–1998. Addiction 2003, 98, 739–747. [Google Scholar]
  42. Kerr, T.; Fairbairn, N.; Tyndall, M.; Marsh, D.; Li, K.; Montaner, J.; Wood, E. Predictors of non-fatal overdose among a cohort of polysubstance-using injection drug users. Drug Alcohol Depend. 2007, 87, 39–45. [Google Scholar] [CrossRef]
  43. Sergeev, B.; Karpets, A.; Sarang, A.; Tikhonov, M. Prevalence and Circumstances of Opiate Overdose Among Injection Drug Users in the Russian Federation. J. Urban Health Bull. N. Y. Acad. Med. 2003, 80, 212–219. [Google Scholar] [CrossRef] [Green Version]
  44. Darke, S.; Duflou, J.; Farrell, M.; Peacock, A.; Lappin, J. Characteristics and circumstances of death related to the self-administration of ketamine. Addiction 2021, 116, 339–345. [Google Scholar] [CrossRef]
  45. Schifano, F. Trapped in the “K-hole”: Overview of deaths associated with ketamine misuse in the UK. J. Clin. Psychopharmacol. 2008, 28, 114–116. [Google Scholar] [CrossRef] [Green Version]
  46. Kalsi, S.S.; Wood, D.M.; Dargan, P.I. The epidemiology and patterns of acute and chronic toxicity associated with recreational ketamine use. Emerg. Health Threat. J. 2011, 4, 7107. [Google Scholar] [CrossRef]
  47. Sen, K.L.R. A review of the nonmedical use of ketamine: Use, users and consequences. J. Psychoact. Drugs 2000, 32, 419–433. [Google Scholar]
  48. Ng, S.; Lee, H.; Chan, Y.; Lau, F. Dilated Common Bile Ducts in Ketamine Abusers. 2009. Available online: www.hkmj.org (accessed on 28 March 2022).
  49. Wong, S.; Lee, K.; Wong, J.; Ng, W.; Cheung, Y.; Lai, P. Dilated Common Bile Ducts Mimicking Choledochal Cysts in Ketamine Abusers. 2009. Available online: www.hkmj.org (accessed on 28 March 2022).
  50. Yu, W.L.; Cho, C.C.M.; Lung, P.F.C.; Hung, E.H.Y.; Hui, J.W.Y.; Chau, H.H.L.; Chan, A.W.; Ahuja, A.T. Ketamine-related cholangiopathy: A retrospective study on clinical and imaging findings. Abdom. Imaging 2014, 39, 1241–1246. [Google Scholar] [CrossRef] [PubMed]
  51. Lo, R.S.; Krishnamoorthy, R.; Freeman, J.G.; Austin, A.S. Cholestasis and biliary dilatation associated with chronic ketamine abuse: A case series. Item Type Article Cholestasis and biliary dilatation associated with chronic ketamine abuse: A case series. Singap. Med. J. 2011, 52, e52–e55. Available online: http://hdl.handle.net/20.500.12904/637 (accessed on 29 March 2022).
  52. Poon, T.L.; Wong, K.F.; Chan, M.Y.; Fung, K.W.; Chu, S.K.; Man, C.W.; You, M.K.; Leung, S.K. Upper gastrointestinal problems in inhalational ketamine abusers. J. Dig. Dis. 2010, 11, 106–110. [Google Scholar] [CrossRef]
  53. Selby, N.M.; Anderson, J.; Bungay, P.; Chesterton, L.J.; Kolhe, N.V. Obstructive nephropathy and kidney injury associated with ketamine abuse. NDT Plus 2008, 1, 310–312. [Google Scholar] [CrossRef] [Green Version]
  54. Wong, G.L.H.; Tam, Y.H.; Ng, C.F.; Chan, A.W.H.; Choi, P.C.L.; Chu, W.C.W.; Lai, P.B.; Chan, H.L.; Wong, V.W. Liver Injury Is Common Among Chronic Abusers of Ketamine. Clin. Gastroenterol. Hepatol. 2014, 12, 1759–1762.e1. [Google Scholar] [CrossRef]
  55. Lee, S.T.; Wu, T.T.; Yu, P.Y.; Chen, R.M. Apoptotic insults to human HepG2 cells induced by S-(+)-ketamine occurs through activation of a Bax-mitochondria-caspase protease pathway. Br. J. Anaesth. 2009, 102, 80–89. [Google Scholar] [CrossRef] [Green Version]
  56. Liu, C.Y.; Xie, D.P.; Liu, J.Z. Microinjection of glutamate into dorsal motor nucleus of the vagus excites gallbladder motility through NMDA receptor—Nitric oxide—cGMP pathway. Neurogastroenterol. Motil. 2004, 16, 347–353. [Google Scholar] [CrossRef]
  57. Rocco, A.; Compare, D.; Angrisani, D.; Sanduzzi Zamparelli Nardone, G. Alcoholic disease: Liver and beyond. World J. Gastroenterol. 2014, 20, 14652–14659. [Google Scholar] [CrossRef]
  58. Seth, D.; Haber, P.S.; Syn, W.K.; Diehl, A.M.; Day, C.P. Pathogenesis of alcohol-induced liver disease: Classical concepts and recent advances. J. Gastroenterol. Hepatol. 2011, 26, 1089–1105. [Google Scholar] [CrossRef]
  59. Mello, T.; Ceni, E.; Surrenti, C.; Galli, A. Alcohol induced hepatic fibrosis: Role of acetaldehyde. Mol. Asp. Med. 2008, 29, 17–21. [Google Scholar] [CrossRef] [PubMed]
  60. Weiner, A.L.; Vieira, L.; Mckay, C.A.; Bayer, M.J. Selected Topics: Toxicology Ketamine Abusers Presenting to The Emergency Department: A Case Series. J. Emerg. Med. 2000, 18, 447–451. [Google Scholar] [CrossRef]
  61. Wai, M.S.; Jiang, Y.; Chan, W.M.; Tsui, T.Y.M.; Tang, H.C.; Lam, W.P.; Fan, M.; Yew, D.T. Long term ketamine and ketamine plus alcohol toxicity—What can we leran from animal models? Mini Rev. Med. Chem. 2013, 13, 273–279. [Google Scholar]
  62. Wei, Y.; Yang, J.; Song, W.; Guo, Q. Gourd-shaped bladder associated with ketamine abuse. Urol. Int. 2012, 89, 123–124. [Google Scholar] [CrossRef] [PubMed]
  63. Turkish, A.; Luo, J.J.; Lefkowitch, J.H. Ketamine abuse, biliary tract disease, and secondary sclerosing cholangitis. Hepatology 2013, 58, 825–827. [Google Scholar] [CrossRef] [PubMed]
  64. Smith, K.M.; Larive, L.L.; Romanelli, F. Club drugs: Methylenedioxymethamphetamine, flunitrazepam, ketamine hydrochloride, and γ-hydroxybutyrate. Am. J. Health-Syst. Pharm. 2022, 59, 1067–1076. Available online: www.clubdrugs.org (accessed on 16 February 2022). [CrossRef] [PubMed]
  65. Kohrs, R.; Durieux, M.E. Ketamine: Teaching an Old Drug New Tricks. Anesth. Analg. 1998, 87, 1186–1193. [Google Scholar]
  66. Yuan, H.; Du, S.; Deng, Y.; Xu, X.; Zhang, Q.; Wang, M.; Wang, P.; Su, Y.; Liang, X.; Sun, Y.; et al. Effects of microRNA-208a on inflammation and oxidative stress in ketamine-induced cardiotoxicity through Notch/NF-κB signal pathways by CHD9. Biosci. Rep. 2019, 39, 1–13. [Google Scholar] [CrossRef] [Green Version]
  67. Mustroph, J.; Lebek, S.; Maier, L.S.; Neef, S. Mechanisms of cardiac ethanol toxicity and novel treatment options. Pharmacol. Ther. 2019, 197, 1–10. [Google Scholar] [CrossRef]
  68. Kinkead, R.; Bach, K.B.; Johnson, S.M.; Hodgeman, B.A.; Mitchell, G.S. Plasticity in respiratory motor control: Intermittent hypoxia and hypercapnia activate opposing serotonergic and noradrenergic modulatory systems. Comp. Biochem. Physiol. Part A 2001, 130, 207–218. [Google Scholar] [CrossRef]
  69. Imam, M.Z.; Kuo, A.; Ghassabian, S.; Smith, M.T. Progress in understanding mechanisms of opioid-induced gastrointestinal adverse effects and respiratory depression. Neuropharmacology 2018, 131, 238–255. [Google Scholar] [CrossRef] [PubMed]
  70. Waters, K.A.; Machaalani, R. Role of NMDA receptors in development of respiratory control. Respir. Physiol. Neurobiol. 2005, 149, 123–130. [Google Scholar] [CrossRef] [PubMed]
  71. Traphagen, N.; Tian, Z.; Allen-Gipson, D. Chronic ethanol exposure: Pathogenesis of pulmonary disease and dysfunction. Biomolecules 2015, 5, 2840–2853. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Chu, P.S.; Ma, W.K.; Yu, C.; Yiu, M.K.; Man, C.W. Destruction of the urinary tract by ketamine abuse: Hong Kong Local experience. SurgPract 2010, 14, 44–48. [Google Scholar] [CrossRef]
  73. Chu, P.S.K.; Ma, W.K.; Wong, S.C.W.; Chu, R.W.H.; Cheng, C.H.; Wong, S.; Tse, J.M.; Lau, F.L.; You, M.K.; Man, C.W. The destruction of the lower urinary tract by ketamine abuse: A new syndrome? BJU Int. 2008, 102, 1616–1622. [Google Scholar] [CrossRef]
  74. Shahani, R.; Streutker, C.; Dickson, B.; Stewart, R.J. Ketamine-Associated Ulcerative Cystitis: A New Clinical Entity. Urology 2007, 69, 810–812. [Google Scholar] [CrossRef]
  75. Rajandram, R.; Yap, N.Y.; Ong, T.A.; Mun, K.S.; Mohamad Wali, H.A.; Hasan, M.S.; Razack, A.H.A.; Ali Mohd, M. Oral ketamine induced pathological changes of the urinary tract in a rat model. Malays. J. Pathol. 2017, 39, 47. [Google Scholar]
  76. Frenkel, C. Urban BW. Molecular actions of racemic ketamine on human cns sodium channels. Br. J. Anaesth. 1992, 69, 292–297. [Google Scholar] [CrossRef]
  77. Han, J.; Kim, N.; Joo, H.; Kim, E. Ketamine blocks Ca 2-activated K channels in rabbit cerebral arterial smooth muscle cells. Am. J. Physiol. -Heart Circ. Physiol. 2003, 285, H1347–H1355. Available online: www.ajpheart.org (accessed on 16 February 2022). [CrossRef]
  78. Kawano, T.; Oshita, S.; Takahashi, A.; Tsutsumi, Y.; Tanaka, K.; Tomiyama, Y.; Kitahata, H.; Nakaya, Y. Molecular Mechanisms Underlying Ketamine-mediated Inhibition of Sarcolemmal Adenosine Triphosphate-sensitive Potassium Channels. Anesthesiology 2005, 102, 93–101. Available online: http://pubs.asahq.org/anesthesiology/article-pdf/102/1/93/357819/0000542-200501000-00017.pdf (accessed on 16 February 2022). [CrossRef]
  79. Chen, X.; Shu, S.; Bayliss, D.A. HCN1 channel subunits are a molecular substrate for hypnotic actions of ketamine. J. Neurosci. 2009, 29, 600–609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Meng, E.; Chang, H.Y.; Chang, S.Y.; Sun, G.H.; Yu, D.S.; Cha, T.L. Involvement of purinergic neurotransmission in ketamine induced bladder dysfunction. J. Urol. 2011, 186, 1134–1141. [Google Scholar] [CrossRef] [PubMed]
  81. Yeung, L.Y.; Rudd, J.A.; Lam, W.P.; Mak, Y.T.; Yew, D.T. Mice are prone to kidney pathology after prolonged ketamine addiction. Toxicol. Lett. 2009, 191, 275–278. [Google Scholar] [CrossRef] [PubMed]
  82. Robinson, D.; Hanna-Mitchell, A.; Rantell, A.; Thiagamoorthy, G.; Cardozo, L. Are we justified in suggesting change to caffeine, alcohol, and carbonated drink intake in lower urinary tract disease? Report from the ICI-RS 2015. Neurourol. Urodyn. 2017, 36, 876–881. [Google Scholar] [CrossRef]
  83. Li, D.; Xu, J.; Liu, F.; Wang, X.; Yang, H.; Li, X. Alcohol Drinking and the Risk of Chronic Kidney Damage: A Meta-Analysis of 15 Prospective Cohort Studies. Alcohol. Clin. Exp. Res. 2019, 43, 1360–1372. [Google Scholar] [CrossRef]
  84. Cheungpasitporn, W.; Thongprayoon, C.; Kittanamongkolchai, W.; Brabec, B.A.; O’corragain, O.A.; Edmonds, P.J.; Erickson, S.B. High alcohol consumption and the risk of renal damage: A systematic review and meta-analysis. QJM 2015, 108, 539–548. [Google Scholar] [CrossRef] [Green Version]
  85. Leal, S.; Ricardo Jorge, D.O.; Joana, B.; Maria, S.S.; Isabel, S.S. Heavy Alcohol Consumption Effects on Blood Pressure and on Kidney Structure Persist after Long-Term Withdrawal. Kidney Blood Press. Res. 2017, 42, 664–675. [Google Scholar] [CrossRef]
  86. Brzóska, M.M.; Moniuszko-Jakoniuk, J.; Piłat-Marcinkiewicz, B.; Sawicki, B. Liver and kidney function and histology in rats exposed to cadmium and ethanol. Alcohol Alcohol. 2003, 38, 2–10. [Google Scholar] [CrossRef] [Green Version]
  87. Husain, K.; Ferder, L.; Ansari, R.A.; Lalla, J. Chronic ethanol ingestion induces aortic inflammation/oxidative endothelial injury and hypertension in rats. Hum. Exp. Toxicol. 2011, 30, 930–939. [Google Scholar] [CrossRef] [Green Version]
  88. Tirapelli, L.F.; Martins-Oliveira, A.; Batalhão, M.E.; Tirapelli, D.P.; Carnio, E.C.; Tanus-Santos, J.E.; Queiroz, R.H.; Padovan, C.M.; Tirapelli, C.R. Ethanol consumption increases the expression of endothelial nitric oxide synthase, inducible nitric oxide synthase and metalloproteinases in the rat kidney. J. Pharm. Pharmacol. 2012, 64, 68–76. [Google Scholar] [CrossRef]
  89. Dinu, D.; Nechifor, M.T.; Movileanu, L. Ethanol-induced alterations of the antioxidant defense system in rat kidney. J. Biochem. Mol. Toxicol. 2006, 19, 386–395. [Google Scholar] [CrossRef] [PubMed]
  90. Hill, G.S.; Heudes, D.; Jacquot, C.; Gauthier, É.; Bariéty, J. Morphometric evidence for impairment of renal autoregulation in advanced essential hypertension. Kidney Int. 2006, 69, 823–831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Ojeda, M.L.; Barrero, M.J.; Nogales, F.; Murillo, M.L.; Carreras, O. Oxidative effects of chronic ethanol consumption on the functions of heart and kidney: Folic acid supplementation. Alcohol Alcohol. 2012, 47, 404–412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Abdulla, M.H.; Johns, E.J. Nitric oxide impacts on angiotensin AT2 receptor modulation of high-pressure baroreflex control of renal sympathetic nerve activity in anaesthetized rats. Acta Physiol. 2014, 210, 832–844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Morgan, C.J.A.; Curran, H.V. Ketamine use: A review. Addiction 2012, 107, 27–38. [Google Scholar] [CrossRef]
  94. Hansen, G.; Jensen, B. The Psychotropic Effect of Ketamine. J. Psychoact. Drugs 1988, 20, 419–425. [Google Scholar] [CrossRef]
  95. Fernandes, L.M.P.; Bezerra, F.R.; Monteiro, M.C.; Silva, M.L.; de Oliveira, F.R.; Lima, R.R.; Fontes-Júnior, E.A.; Maia, C.S.F. Thiamine deficiency, oxidative metabolic pathways and ethanol-induced neurotoxicity: How poor nutrition contributes to the alcoholic syndrome, as Marchiafava-Bignami disease. Eur. J. Clin. Nutr. Nat. Publ. Group 2017, 71, 580–5866. [Google Scholar] [CrossRef]
  96. Fernandes, L.M.P.; de Andrade, E.F.; Monteiro, M.C.; Cartágenes, S.C.; Lima, R.R.; Prediger, R.D.; Maia, C.S.F. Ethanol: Neurotoxicity and Brain Disorders. In Addictive Substances and Neurological Disease: Alcohol, Tobacco, Caffeine, and Drugs of Abuse in Everyday Lifestyles; Elsevier: Amsterdam, The Netherlands, 2017; pp. 201–215. [Google Scholar]
  97. Morgan, C.J.A.; Curran, H.V. Acute and chronic effects of ketamine upon human memory: A review. Psychopharmacology 2006, 188, 408–424. [Google Scholar] [CrossRef]
  98. Stefanovic, A.; Brandner, B.; Klaassen, E.; Cregg, R.; Nagaratnam, M.; Bromley, L.M.; Das, R.K.; Rossell, S.L.; Morgan, C.J.; Curran, H.V. Acute and chronic effects of ketamine on semantic priming: Modeling schizophrenia. J. Clin. Psychopharmacol. 2009, 29, 124–133. [Google Scholar] [CrossRef]
  99. Edward Roberts, R. Abnormalities in white matter microstructure associated with chronic ketamine. Neuropsychopharmacology 2014, 39, 329–338. [Google Scholar] [CrossRef] [Green Version]
  100. Blagrove, M.; Morgan, C.J.A.; Curran, H.V.; Bromley, L.; Brandner, B. The incidence of unpleasant dreams after sub-anaesthetic ketamine. Psychopharmacology 2009, 203, 109–120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Muetzelfeldt, L.; Kamboj, S.K.; Rees, H.; Taylor, J.; Morgan, C.J.A.; Curran, H.V. Journey through the K-hole: Phenomenological aspects of ketamine use. Drug Alcohol Depend. 2008, 95, 219–229. [Google Scholar] [CrossRef] [PubMed]
  102. Morgan, C.J.A.; Duffin, S.; Hunt, S.; Monaghan, L.; Mason, O.; Curran, H.V. Neurocognitive function and schizophrenia-proneness in individuals dependent on ketamine, on high potency cannabis (skunk) or on cocaine. Pharmacopsychiatry 2012, 45, 269–274. [Google Scholar] [CrossRef]
  103. Morgan, C.J.A.; Mofeez, A.; Brandner, B.; Bromley, L.; Curran, H.V. Acute Effects of Ketamine on Memory Systems and Psychotic Symptoms in Healthy Volunteers. Neuropsychopharmacology 2004, 29, 208–218. [Google Scholar] [CrossRef] [PubMed]
  104. Morgan, C.J.A.; Riccelli, M.; Maitland, C.H.; Curran, H.V. Long-term effects of ketamine: Evidence for a persisting impairment of source memory in recreational users. Drug Alcohol Depend. 2004, 75, 301–308. [Google Scholar] [CrossRef]
  105. Morgan, C.J.A.; Monaghan, L.; Curran, H.V. Beyond the K-hole: A 3-year longitudinal investigation of the cognitive and subjective effects of ketamine in recreational users who have substantially reduced their use of the drug. Addiction 2004, 99, 1450–1461. [Google Scholar] [CrossRef]
  106. Mason, O.J.; Morgan, C.J.M.; Stefanovic, A.; Curran, H.V. The Psychotomimetic States Inventory (PSI): Measuring psychotic-type experiences from ketamine and cannabis. Schizophr. Res. 2008, 103, 138–142. [Google Scholar] [CrossRef]
  107. Freeman, T.P.; Morgan, C.J.A.; Klaassen, E.; Das, R.K.; Stefanovic, A.; Brandner, B.; Curran, H.V. Superstitious conditioning as a model of delusion formation following chronic but not acute ketamine in humans. Psychopharmacology 2009, 206, 563–573. [Google Scholar] [CrossRef]
  108. Adell, A. Brain NMDA receptors in schizophrenia and depression. Biomolecules 2020, 10, 947. [Google Scholar] [CrossRef]
  109. Shelley, A.M.; Silipo, G.; Javitt, D.C. Diminished responsiveness of ERPs in schizophrenic subjects to changes in auditory stimulation parameters: Implications for theories of cortical dysfunction. Schizophr. Res. 1999, 37, 65–79. [Google Scholar] [CrossRef]
  110. Stone, J.M.; Erlandsson, K.; Arstad, E.; Squassante, L.; Teneggi, V.; Bressan, R.A.; Krystal, J.H.; Ell, P.J.; Pilowsky, L.S. Relationship between ketamine-induced psychotic symptoms and NMDA receptor occupancy—A [123I]CNS-1261 SPET study. Psychopharmacology 2008, 197, 401–408. [Google Scholar] [CrossRef]
  111. Hakami, T.; Jones, N.C.; Tolmacheva, E.A.; Gaudias, J.; Chaumont, J.; Salzberg, M.; O’Brien, T.J.; Pinault, D. NMDA receptor hypofunction leads to generalized and persistent aberrant γ oscillations independent of hyperlocomotion and the state of consciousness. PLoS ONE 2009, 4, e6755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Lee, K.H.; Williams, L.; Haig, A.; Gordon, E. “Gamma (40 Hz) phase synchronicity” and symptom dimensions in schizophrenia. Cogn. Neuropsychiatry 2003, 8, 57–71. [Google Scholar] [CrossRef] [PubMed]
  113. Sullivan, E.M.; Timi, P.; Hong, L.E.; O’Donnell, P. Reverse translation of clinical electrophysiological biomarkers in behaving rodents under acute and chronic NMDA receptor antagonism. Neuropsychopharmacology 2015, 40, 719–727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Abdallah, C.G.; de Feyter, H.M.; Averill, L.A.; Jiang, L.; Averill, C.L.; Chowdhury, G.M.I.; Purohit, P.; de Graaf, R.A.; Esterlis, I.; Juchem, C.; et al. The effects of ketamine on prefrontal glutamate neurotransmission in healthy and depressed subjects. Neuropsychopharmacology 2018, 43, 2154–2160. [Google Scholar] [CrossRef]
  115. Callicott, J.H.; Bertolino, A.; Mattay, V.S.; Langheim, F.J.; Duyn, J.; Coppola, R.; Goldberg, T.E.; Weinberger, D.R. Physiological dysfunction of the dorso lateral prefrontal cortex in schizophrenia revisited. Cereb. Cortex. 2000, 10, 1078–1092. [Google Scholar] [CrossRef]
  116. Dienel, S.J.; Enwright, J.F.; Hoftman, G.D.; Lewis, D.A. Markers of glutamate and GABA neurotransmission in the prefrontal cortex of schizophrenia subjects: Disease effects differ across anatomical levels of resolution. Schizophr. Res. 2020, 217, 86–94. [Google Scholar] [CrossRef]
  117. Lathi, A.C.; Holcomb, H.H.; Medoff, D.R.; Tamminga, C.A. Ketamine activates psychosis and alters limbic blood flow in schizophrenia. Neuroreport 1995, 6, 869–872. [Google Scholar]
  118. Kondziella, D.; Brenner, E.; Eyjolfsson, E.M.; Markinhuhta, K.R.; Carlsson, M.L.; Sonnewald, U. Glial-neuronal interactions are impaired in the schizophrenia model of repeated MK801 exposure. Neuropsychopharmacology 2006, 31, 1880–1887. [Google Scholar] [CrossRef]
  119. Vollenweider, F.X.; Vontobel, P.; Éye, I.; Hell, D.; Leenders, K.L. Effects of (S)-ketamine on Striatal Dopamine: A [11 C]raclopride PET Study of a Model Psychosis in Humans. 2002. Available online: www.elsevier.com/locate/jpsychires (accessed on 16 February 2022).
  120. Martin, P.; CMHS. Systemic PCP treatment elevates brain extracellular 5-HT: A microdialysis study in awake rats. Neuroreport 1998, 9, 2985–2988. [Google Scholar] [CrossRef]
  121. Nelson, C.L.; Burk, J.A.; Bruno, J.P.; Sarter, M. Effects of acute and repeated systemic administration of ketamine on prefrontal acetylcholine release and sustained attention performance in rats. Psychopharmacology 2002, 161, 168–179. [Google Scholar] [CrossRef] [PubMed]
  122. Pal, D.; Hambrecht-Wiedbusch, V.S.; Silverstein, B.H.; Mashour, G.A. Electroencephalographic coherence and cortical acetylcholine during ketamine-induced unconsciousness. Br. J. Anaesth. 2015, 114, 979–989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Seeman, P.; Ko, F.; Tallerico, T. Dopamine receptor contribution to the action of PCP, LSD and ketamine psychotomimetics. Mol. Psychiatry 2005, 10, 877–883. [Google Scholar] [CrossRef] [PubMed]
  124. Kapur, S.; Seeman, P. NMDA receptor antagonists ketamine and PCP have direct effects on the dopamine D2 and serotonin 5-HT2 receptors—Implications for models of schizophrenia. Mol. Psychiatry 2002, 7, 837–844. [Google Scholar] [CrossRef] [Green Version]
  125. Kinoshita, H.; Nishitani, N.; Nagai, Y.; Andoh, C.; Asaoka, N.; Kawai, H.; Shibui, N.; Nagayasu, K.; Shirakawa, H.; Nakagawa, T.; et al. Ketamine-Induced Prefontal Serotonin Realease Is Mediated by Cholinergic Neurons in the Penduculopontine Tegmental Nucleus. Int. J. Neuropsychopharmacol. 2018, 21, 305–310. [Google Scholar] [CrossRef] [Green Version]
  126. Frohlich, J.; van Horn, J.D. Reviewing the ketamine model for schizophrenia. J. Psychopharmacol. 2014, 28, 287–302. [Google Scholar] [CrossRef]
  127. Shibuya, H.; Mori, H.; Toru, M. Sigma Receptors in Schizophrenic Cerebral Cortices*. Neurochem. Res. 1992, 17, 983–990. [Google Scholar] [CrossRef]
  128. Weissman, A.D.; Casanova, M.F.; Kleinman, J.E.; London, E.D.; de Souza, E.B. Selective Loss of Cerebral Cortical Sigma, but Not PCP Binding Sites in Schizophrenia. Biol. Psychiatry 1991, 29, 41–54. [Google Scholar] [CrossRef]
  129. Pfeiffer, A.; Branti, V.; Herz, A.; Emrich, H.M. Psychotomimesis Mediated by K Opiate Receptors. Science 1986, 233, 774. [Google Scholar] [CrossRef]
  130. Tejeda, H.A.; Shippenberg, T.S.; Henriksson, R. The dynorphin/κ-opioid receptor system and its role in psychiatric disorders. Cell. Mol. Life Sci. 2012, 69, 857–896. [Google Scholar] [CrossRef]
  131. Marcel, L. De la Folie Causée par L’abus dês Boissons Alcooliques. Ph.D. Thesis, Imp. Rignoux, Paris, France, 1847. [Google Scholar]
  132. Soyka, M. 1990. Psychopathological characteristics in alcohol hallucinosis and paranoid schizophrenia. Acta Psychiatr. Scand. 1990, 81, 255–259. [Google Scholar] [CrossRef] [PubMed]
  133. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 4th ed.; American Psychiatric Association: Washington, DC, USA, 2000. [Google Scholar]
  134. Glass, I.B. Alcoholic Hallucinosis: A psychiatric enigma-1. The Development of an Idea. Br. J. Addict. 1989, 84, 29–41. [Google Scholar] [CrossRef] [PubMed]
  135. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders; American Psychiatric Association: Washington, DC, USA, 1980. [Google Scholar]
  136. Kitabayashi, Y.; Narumoto, J.; Shibata, K.H.; Fukui, K. Neuropsychiatric Background of Alcohol Hallucinosis: A Spect Study. J. Neuropsychiatry 2007, 19, 85. [Google Scholar] [CrossRef] [PubMed]
  137. Soyka, M.; ZTDSTK. FDG-PET and IBZMSPECT suggest reduced thalamic activity but no dopaminergic dysfunction in chronic alcohol hallucinosis. J. Neuropsychiatry Clin. Neurosci. 2000, 12, 287–288. [Google Scholar] [CrossRef]
  138. Aliyev, N.A.; Aliyev, Z.N. Application of glycine in acute alcohol hallucinosis. Hum. Psychopharmacol. 2005, 20, 591–594. [Google Scholar] [CrossRef]
  139. Branchey, L.; Branchey, M.; Worner, T.M.; Zucker, D.; Shaw, S.; Lieber, C.S. Association between Amino Acid Alterations and Hallucinations in Alcoholic Patients. Biol. Psychiatry 1985, 20, 1167–1173. [Google Scholar] [CrossRef]
  140. Fadda, F.; Mosca, E.; Colombo, G.; Gessa, G.L. Effect of spontaneous ingestion of ethanol on brain dopamine metabolism. Life Sci. 1989, 44, 281–287. [Google Scholar] [CrossRef]
  141. Zuo, D.; Liu, Y.; Liu, Z.; Cui, J.; Zhou Liu, Y.; Li, Z.; Wu, Y. Alcohol aggravates ketamine-induced behavioral, morphological and neurochemical alterations in adolescent rats: The involvement of CREB-related pathways. Behav. Brain Res. 2018, 349, 80–90. [Google Scholar] [CrossRef]
  142. Liu, Y.; Lin, D.; Wu, B.; Zhou, W. Ketamine abuse potential and use disorder. Brain Res. Bull. 2016, 126, 68–73. [Google Scholar] [CrossRef] [Green Version]
  143. Tsai, G.; Gastfriend, D.R.; Coyle, J.T. The Glutamatergic Basis of Human Alcoholism. Am. J. Psychiatry 1995, 152, 332–340. [Google Scholar]
  144. Vengeliene, V.; Bilbao, A.; Molander, A.; Spanagel, R. Neuropharmacology of alcohol addiction. Br. J. Pharmacol. 2008, 154, 299–315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Jung, M.E.; Metzger, D.B. Alcohol withdrawal and brain injuries: Beyond classical mechanisms. Molecules 2010, 15, 4984–5011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Viviani, B.; Boraso, M.; Marchetti, N.; Marinovich, M. Perspectives on neuroinflammation and excitotoxicity: A neurotoxic conspiracy? NeuroToxicology 2014, 43, 10–20. [Google Scholar] [CrossRef]
  147. Miladinovic, T.; Nashed, M.G.; Singh, G. Overview of glutamatergic dysregulation in central pathologies. Biomolecules 2015, 5, 3112–3141. [Google Scholar] [CrossRef] [Green Version]
  148. Ménard, C.; Hodes, G.E.; Russo, S.J. Pathogenesis of depression: Insights from human and rodent studies. Neuroscience 2016, 321, 138–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Beurel, E.; Toups, M.; Nemeroff, C.B. The Bidirectional Relationship of Depression and Inflammation: Double Trouble. Neuron 2020, 107, 234–256. [Google Scholar] [CrossRef]
  150. Réus, G.Z.; Fries, G.R.; Stertz, L.; Badawy, M.; Passos, I.C.; Barichello, T.; Kapczinski, F.; Quevedo, J. The role of inflammation and microglial activation in the pathophysiology of psychiatric disorders. Neuroscience 2015, 300, 141–154. [Google Scholar] [CrossRef] [PubMed]
  151. Volkow, N.D.; Michaelides, M.; Baler, R. The neuroscience of drug reward and addiction. Physiol. Rev. 2019, 99, 2115–2140. [Google Scholar] [CrossRef]
  152. Baik, J.H. Dopamine signaling in reward-related behaviors. Front. Neural Circuits 2013, 7, 152. [Google Scholar] [CrossRef] [Green Version]
  153. Lovinger, D.M. Excitotoxicity alcohol—Related Brain Damage. Alcohol ClinExp Res. 1993, 17, 19–27. [Google Scholar] [CrossRef]
  154. Schuckit, M.A.; Hesseibrock, V. Alcohol Dependence and Anxiety Disorders: What Is the Relationship? Focus 1994, 151, 1723–1734. [Google Scholar] [CrossRef]
  155. Yang, J.Y.; Xue, X.; Tian, H.; Wang, X.X.; Dong, Y.X.; Wang, F.; Zhao, Y.N.; Yao, X.C.; Cui, W.; Wu, C.F. Role of microglia in ethanol-induced neurodegenerative disease: Pathological and behavioral dysfunction at different developmental stages. Pharmacol. Ther. 2014, 144, 321–337. [Google Scholar] [CrossRef] [PubMed]
  156. Rodríguez-Rabassa, M.; López, P.; Sánchez, R.; Hernández, C.; Rodríguez, C.; Rodríguez-santiago, R.E.; Orengo, J.C.; Green, V.; Yamamura, Y.; Rivera-Amill, V. Inflammatory biomarkers, microbiome, depression, and executive dysfunction in alcohol users. Int. J. Environ. Res. Public Health 2020, 17, 689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Belém-Filho, I.J.A.; Ribera, P.C.; Nascimento, A.L.; Gomes, A.R.Q.; Lima, R.R.; Crespo-Lopez, M.E.; Monteiro, M.C.; Fontes-Júnior, E.A.; Lima, M.O.; Maia, C.S.F. Low doses of methylmercury intoxication solely or associated to ethanol binge drinking induce psychiatric-like disorders in adolescent female rats. Environ. Toxicol. Pharmacol. 2018, 60, 184–194. [Google Scholar] [CrossRef]
  158. Clayton, C.J.; Hicks, R.E. Ethanol, monoamines, and affect. J. Neural Transm. 1994, 98, 169–195. [Google Scholar] [CrossRef]
  159. Milio, C.; Hadfield, M.G. Ethanol Alters Monoamines in Mouse Brain Regions Specific. Brain Res. Bull. 1992, 29, 599–603. [Google Scholar] [CrossRef]
  160. Kaneyuki, T.; Morimasa, T.; Okada, H.; Shohmori, T. Acta Medica Okayama the effect of acute and repeated ethanol administration on monoamines and their metabolites in brain regions of rats. Acta Med. Okayama 1991, 45, 201–208. [Google Scholar]
  161. Costardi, J.V.V.; Nampo, R.A.T.; Silva, G.L.; FerreiraRibeiro, M.A.; Stella, H.J.; Stella, M.B.; Malheiros, S.V. A review on alcohol: From the central action mechanism to chemical dependency. Rev. Assoc. Med. Bras. 2015, 61, 381–387. [Google Scholar] [CrossRef] [Green Version]
  162. Dixon, M.L.; Thiruchselvam, R.; Todd, R.; Christoff, K. Emotion and the prefrontal cortex: An integrative review. Psychol. Bull. 2017, 143, 1033–1081. [Google Scholar] [CrossRef]
  163. Esperidião-Antonio, V.; Majeski-Colombo, M.; Toledo-Monteverde, D.; Moraes-Martins, G.; Fernandes, J.J.; Bauchiglioni de Assis, M.; Montenegro, S.; Siqueira-Batista, R. Neurobiology of emotions: An update. Int. Rev. Psychiatry 2017, 29, 293–307. [Google Scholar] [CrossRef]
  164. Li, Q.; Wu, H.R.; Fan, S.J.; Liu, D.X.; Jiang, H.; Zhang, Q.; Pan, F. The effects of sub-anesthetic ketamine plus ethanol behaviors and apoptosis in the prefrontal cortex and hippocampus of adolescent rats. Pharmacol. Biochem. Behav. 2019, 184, 1727–1742. [Google Scholar] [CrossRef] [PubMed]
  165. Fan, N.; Xu, K.; Ning, Y.; Rosenheck, R.; Wang, D.; Ke, X.; Ding, Y.; Sun, B.; Zhou, C.; Deng, X.; et al. Profiling the psychotic, depressive and anxiety symptoms in chronic ketamine users. Psychiatry Res. 2016, 237, 311–315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Zhang, Y.; Xu, Z.; Zhang, S.; Desrosiers, A.; Schottenfeld, R.S.; Chawarski, M.C. Profiles of psychiatric symptoms among amphetamine type stimulant and ketamine using inpatients in Wuhan, China. J. Psychiatr. Res. 2014, 53, 99–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Silote, G.P.; de Oliveira, S.F.S.; Ribeiro, D.E.; Machado, M.S.; Andreatini, R.; Joca, S.R.L.; Beijamini, V. Ketamine effects on anxiety and fear-related behaviors: Current literature evidence and new findings. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2020, 100, 109878. [Google Scholar] [CrossRef] [PubMed]
  168. Perez-Caballero, L.; Torres-Sanchez, S.; Romero-López-Alberca, C.; González-Saiz, F.; Mico, J.A.; Berrocoso, E. Monoaminergic system and depression. Cell Tissue Res. 2019, 377, 107–113. [Google Scholar] [CrossRef] [PubMed]
  169. Nicolaides, N.C.; Kyratzi, E.; Lamprokostopoulou, A.; Chrousos, G.P.; Charmandari, E. Stress, the stress system and the role of glucocorticoids. NeuroImmunoModulation 2014, 22, 6–19. [Google Scholar] [CrossRef]
  170. Ann Stephens, M.; Wand, G. Stress and the HPA Axis Role of Glucocorticoids in Alcohol Dependence. Alcohol Res. Curr. Rev. 2012, 4, 468–483. [Google Scholar]
  171. Allen, D.C.; Gonzales, S.W.; Grant, K.A. Effect of repeated abstinence on chronic ethanol self-administration in the rhesus monkey. Psychopharmacology 2018, 235, 109–120. [Google Scholar] [CrossRef]
  172. Rivier, C.; Imaki, T. Prolonged exposure to alcohol: Effect on CRF mRNA levels, and CRF-and stress-induced ACTH secretion in the rat. Brain Res. 1990, 520, 1–5. [Google Scholar] [CrossRef]
  173. Silvestre, J.S.; Pallarés, M.; Nadal, R.; Ferré, N. Opposite Effetc of Ethanol and Ketamine the Elevated plus-maze test Wistar rats undergoing a chronic oral voluntary consumption procedure. J. Psychopharmacol. 2002, 16, 305–312. [Google Scholar] [CrossRef]
  174. Sampaio, T.B.; de Oliveira, L.F.; Constantino, L.C.; Costa, A.P.; Poluceno, G.G.; Martins, W.C.; Dal-Cim, T.; de Oliveira, K.A.; Ludka, F.K.; Prediger, R.D.; et al. Long-Term Neurobehavioral Consequences of a Single Ketamine Neonatal Exposure in Rats: Effects on Cellular Viability and Glutamate Transport in Frontal Cortex and Hippocampus. Neurotox. Res. 2018, 34, 649–659. [Google Scholar] [CrossRef] [PubMed]
  175. Machado-Ferraro, K.M.; Soriano-de-Mello, D.S.; de Moura, I.P.; da Silveira, C.C.S.M.; de Farias, E.C.F.; Maia, M.L.F.; de Sales, S.C.D.; Carvalho, A.E.V.; Magno, I.M.N.; Fontes-Júnior, E.A.; et al. Long-lasting neurocognitive disorders: A case report of previously undescribed adverse effects after ketamine sedation and analgesia in a pediatric patient. Ann. Transl. Med. 2022, 10, 113. [Google Scholar] [CrossRef] [PubMed]
  176. Lisman, J.; Cooper, K.; Sehgal, M.; Silva, A.J. Memory formation depends on both synapse-specific modifications of synaptic strength and cell-specific increases in excitability. Nat. Neurosci. 2018, 21, 309–3014. [Google Scholar] [CrossRef] [Green Version]
  177. Asok, A.; Leroy, F.; Rayman, J.B.; Kandel, E.R. Molecular Mechanisms of the Memory Trace. Trends Neurosci. 2019, 42, 14–22. [Google Scholar] [CrossRef] [PubMed]
  178. Colucci-D’amato, L.; Speranza, L.; Volpicelli, F. Neurotrophic factor bdnf, physiological functions and therapeutic potential in depression, neurodegeneration and brain cancer. Int. J. Mol. Sci. 2020, 21, 1–29. [Google Scholar]
  179. Itoh, N.; Enomoto, A.; Nagai, T.; Takahashi, M.; Yamada, K. Molecular mechanism linking BDNF/TrkB signaling with the NMDA receptor in memory: The role of Girdin in the CNS. Rev. Neurosci. 2016, 27, 481–490. [Google Scholar] [CrossRef]
  180. Ortega-Martínez, S. A new perspective on the role of the CREB family of transcription factors in memory consolidation via adult hippocampal neurogenesis. Front. Mol. Neurosci. 2015, 8, 46. [Google Scholar] [CrossRef] [Green Version]
  181. Narendran, R.; Gordon Frankle, W.; Keefe, R.; Gil, R.; Martinez, D.; Slifstein, M.; Kegeles, L.S.; Talbot, P.S.; Huang, Y.; Hwang, D.R.; et al. Altered Prefrontal Dopaminergic Function in Chronic Recreational Ketamine Users. Am. J. Psychiatry 2005, 162, 2352–2359. Available online: http://ajp.psychiatryonline.org (accessed on 16 February 2022). [CrossRef] [Green Version]
  182. Brust, J.C.M. Ethanol and cognition: Indirect effects, neurotoxicity and neuroprotection: A review. Int. J. Environ. Res. Public Health 2010, 7, 1540–1557. [Google Scholar] [CrossRef]
  183. Pfisterer, U.; Khodosevich, K. Neuronal survival in the brain: Neuron type-specific mechanisms. Cell Death Dis. 2017, 8, e2643. [Google Scholar] [CrossRef]
  184. Reichardt, L.F. Neurotrophin-regulated signalling pathways. Philos. Trans. R. Soc. B Biol. Sci. 2006, 361, 1545–1564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Zhang, J.; Wang, Y.; Liu, X.; Dagda, R.K.; Zhang, Y. How AMPK and PKA interplay to regulate mitochondrial function and survival in models of ischemia and diabetes. Oxid. Med. Cell. Longev. 2017, 2017, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Kamat, P.K.; Kalani, A.; Rai, S.; Swarnkar, S.; Tota, S.; Nath, C.; Tyagi, N. Mechanism of Oxidative Stress and Synapse Dysfunction in the Pathogenesis of Alzheimer’s Disease: Understanding the Therapeutics Strategies. Mol. Neurobiol. 2016, 53, 648–661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Wang, R.; Reddy, P.H. Role of Glutamate and NMDA Receptors in Alzheimer’s Disease. J. Alzheimer’s Dis. 2017, 57, 1041–1048. [Google Scholar] [CrossRef] [Green Version]
  188. Zádori, D.; Veres, G.; Szalárdy, L.; Klivényi, P.; Vécsei, L. Alzheimer’s Disease: Recent Concepts on the Relation of Mitochondrial Disturbances, Excitotoxicity, Neuroinflammation, and Kynurenines. J. Alzheimer’s Dis. 2018, 62, 523–547. [Google Scholar] [CrossRef] [Green Version]
  189. Aleksandrova, L.R.; Wang, Y.T.; Phillips, A.G. Hydroxynorketamine: Implications for the NMDA Receptor Hypothesis of Ketamine’s Antidepressant Action. Chronic Stress 2017, 1, 2470547017743511. [Google Scholar] [CrossRef] [Green Version]
  190. Hashimoto, K. Molecular mechanisms of the rapid-acting and long-lasting antidepressant actions of (R)-ketamine. Biochem. Pharmacol. 2020, 177, 113935. [Google Scholar] [CrossRef]
  191. Brum, G.F.; Rosa, H.Z.; Rossato, D.R.; Rosa, J.L.O.; Metz, V.G.; Milanesi, L.H.; Burger, M.E. Binge and Subchronic Exposure to Ketamine Promote Memory Impairments and Damages in the Hippocampus and Peripheral Tissues in Rats: Gallic Acid Protective Effects. Neurotox. Res. 2020, 38, 274–286. [Google Scholar] [CrossRef]
  192. Kunitoh, S.; Tanaka, T.; Imaoka, S.; Funaet, Y.; Monna, Y. Elsevier Science Ltd Medical Council on Alcoholism contribution of cytochrome p450s to meos (microsomal ethanol-oxidizing system): A specific and sensitive assay of meos activity by hplc with fluorescence labeling. Pergamon Alcohol Alcohol. 1993, 28, 63–68. [Google Scholar] [CrossRef]
  193. Kamp, J.; Jonkman, K.; van Velzen, M.; Aarts, L.; Niesters, M.; Dahan, A.; Olofsen, E. Pharmacokinetics of ketamine and its major metabolites norketamine, hydroxynorketamine, and dehydronorketamine: A model-based analysis. Br. J. Anaesth. 2020, 125, 750–761. [Google Scholar] [CrossRef]
  194. Mladěnka, P.; Applová, L.; Patočka, J.; Costa, V.M.; Remiao, F.; Pourová, J.; Mladěnka, A.; Karlíčková, J.; Jahodář, L.; Vopršalová, M.; et al. Comprehensive review of cardiovascular toxicity of drugs and related agents. Med. Res. Rev. 2018, 38, 1332–1403. [Google Scholar] [CrossRef] [PubMed]
  195. Suhail, M. Na+, K+-ATPase: Ubiquitous Multifunctional Transmembrane Protein and its Relevance to Various Pathophysiological Conditions. J. Clin. Med. Res. 2010, 2, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Bhatia, D.; Capili, A.; Choi, M.E. Mitochondrial dysfunction in kidney injury, inflammation, and disease: Potential therapeutic approaches. Kidney Res. Clin. Pract. 2020, 39, 244–258. [Google Scholar] [CrossRef] [PubMed]
  197. Patergnani, S.; Bouhamida, E.; Leo, S.; Pinton, P.; Rimessi, A. Mitochondrial oxidative stress and “mito-inflammation”: Actors in the diseases. Biomedicines 2021, 9, 216. [Google Scholar] [CrossRef]
  198. Strong, C.E.; Kabbaj, M. Neural Mechanisms Underlying the Rewarding and Therapeutic Effects of Ketamine as a Treatment for Alcohol Use Disorder. Front. Behav. Neurosci. 2020, 14, 593860. [Google Scholar] [CrossRef]
  199. Olsen, R.W.; Liang, J. Role of GABAA receptors in alcohol use disorders suggested by chronic intermittent ethanol (CIE) rodent model Tim Bliss. Mol. Brain 2017, 10, 1–20. [Google Scholar] [CrossRef]
  200. Nguyen, D.; Alavi, M.V.; Kim, K.Y.; Kang, T.; Scott, R.T.; Noh, Y.H.; Lindsey, J.D.; Wissinger, B.; Ellisman, M.H.; Weinreb, R.N.; et al. A new vicious cycle involving glutamate excitotoxicity, oxidative stress and mitochondrial dynamics. Cell Death Dis. 2011, 2, e240. [Google Scholar] [CrossRef]
  201. Tapia-Rojas, C.; Pérez, M.J.; Jara, C.; Vergara, E.H.; Quintanilla, R.A. Ethanol Consumption Affects Neuronal Function: Role of the Mitochondria. Mitochondrial Dis. 2017. Available online: https://www.intechopen.com/chapters/58876 (accessed on 26 June 2022).
  202. Oliveira, A.C.A.; Pereira, M.C.S.; Santana, L.N.D.S.; Fernandes, R.M.; Teixeira, F.B.; Oliveira, G.B.; Fernandes, L.M.; Fontes-Júnior, E.A.; Prediger, R.D.; Crespo-López, M.E.; et al. Chronic ethanol exposure during adolescence through early adulthood in female rats induces emotional and memory deficits associated with morphological and molecular alterations in hippocampus. J. Psychopharmacol. 2015, 29, 712–724. [Google Scholar] [CrossRef]
Figure 1. Schematic pharmacological targets with potential synergism between deleterious effects of ketamine (red markers) and ethanol (black markers), highlighting (A) the main clinical manifestations already described and effects related to (B) hepatic, (C) cardiac, (D) respiratory, and (E) urinary damage. (+) Activation, elevation, potentiation, or stimulation; (−) inhibition or reduction; (X) blocking; (✸) damage or disruption.
Figure 1. Schematic pharmacological targets with potential synergism between deleterious effects of ketamine (red markers) and ethanol (black markers), highlighting (A) the main clinical manifestations already described and effects related to (B) hepatic, (C) cardiac, (D) respiratory, and (E) urinary damage. (+) Activation, elevation, potentiation, or stimulation; (−) inhibition or reduction; (X) blocking; (✸) damage or disruption.
Ijms 23 07800 g001
Figure 2. Principal pharmacological mechanisms and repercussions of ketamine (red markers) or ethanol (black markers) use during drugs bioavailability on (A) glutamatergic, GABAergic, (B) dopaminergic, serotoninergic, cholinergic, and opioidergic neurotransmission on the central nervous system. (+) Activation, elevation, potentiation, or stimulation; (−) inhibition or reduction; (X) blocking.
Figure 2. Principal pharmacological mechanisms and repercussions of ketamine (red markers) or ethanol (black markers) use during drugs bioavailability on (A) glutamatergic, GABAergic, (B) dopaminergic, serotoninergic, cholinergic, and opioidergic neurotransmission on the central nervous system. (+) Activation, elevation, potentiation, or stimulation; (−) inhibition or reduction; (X) blocking.
Ijms 23 07800 g002
Figure 3. Principal repercussions of ketamine (red markers) or ethanol (black markers) withdrawal on (A) glutamatergic, GABAergic, (B) dopaminergic, and serotoninergic neurotransmission on the central nervous system. (✸) Damage or disruption.
Figure 3. Principal repercussions of ketamine (red markers) or ethanol (black markers) withdrawal on (A) glutamatergic, GABAergic, (B) dopaminergic, and serotoninergic neurotransmission on the central nervous system. (✸) Damage or disruption.
Ijms 23 07800 g003
Table 1. Effects reported in studies about ketamine and ethanol solely or combined on cardiovascular, respiratory, urinary, and hepatobiliary systems.
Table 1. Effects reported in studies about ketamine and ethanol solely or combined on cardiovascular, respiratory, urinary, and hepatobiliary systems.
Drug(s)Evaluation ConditionStudy
Information
Organ(s) or Body(s) System(s)Main Effects DescribedPossible MechanismsReference
KetamineUnder drug effectsLiterature reviewCardiovascular and urinaryAcute: hypertension and tachycardia.
Chronic: risk of hemorrhagic/ulcerative cystitis and obstructive nephropathy
Not investigated[46]
KetamineUnder drug effectsPattern of use: chronic
Type: clinical study
Finality of use:
recreational
Dose and use
frequency:
not informed
Liver, biliary,
urinary, and
cardiorespiratory system
Under drug: cholestasis and biliary dilatation;
hypotension; tachycardia and tachypnoea; bilateral hydronephrosis; acute
renal failure; and increase in hepatic transaminases and alkaline phosphatase
Not investigated[51]
KetamineUnder drug effectsPattern of use: acute
Type: pre-clinical study
Finality of use: not informed
Dose and use
frequency: 10, 50, 100, and 200 mM for 1, 6, or 24 h
Liver (in vitro)Apoptosis and decreased cell viabilityDNA fragmentation, mitochondrial membrane potential and adenosine triphosphate levels decrease;
cytosolic cytochrome and caspase-9, -3, and -6 activities increase
[55]
KetamineUnder drug effectsPattern of use: acute
Type: pre-clinical study
Finality of use: not informed
Dose and use
frequency: a single administration of 180 mmol/L
HepatobiliaryStrength of phasic gallbladder contraction
decrease
NMDA blockade?[56]
KetamineWithdrawal and drug
effects
Pattern of use: chronic
Type: clinical study
Finality of use:
recreational
Dose and use
frequency:
not informed
Urinary systemIntractable dysuria,
painful urination, and gross hematuria
Not investigated[62]
KetamineUnder drug effectsPattern of use: chronic
Type: clinical study
Finality of use:
recreational
Dose and use
frequency:
frequency not
informed; dose 100–200 mg (i.m.)/ intranasal route dose not informed
Cardiovascular systemChest pain, palpitations, tachycardia, and hypertensionNot investigated[60]
KetamineUnder drug and
withdrawal effects
Literature reviewCardiorespiratory systemHypertension, tachycardia, and palpitations;
respiratory toxicity,
including depression and apnea
Authors propose that cardiovascular toxicity can result from reflex sympathetic activation[64]
KetamineUnder drug effectsPattern of use: acute
Type: pre-clinical study
Finality of use: not informed
Dose and use
frequency: single dose of 100 mg (i.p.)
Cardiovascular systemCardiotoxicityIncrease of microRNA-208a, accompanied by
increased inflammation and oxidative stress, suppression of CHD9 and Notch1, and induction of p65
protein expression in vitro model
[66]
KetamineNot informedPattern of use: not specified
Type: clinical study
Finality of use:
recreational
Dose and use
frequency:
not informed
HepatobiliaryDilated common bile ducts and increase of alkaline phosphataseNot investigated[48]
KetamineNot informedLiterature reviewUrinary systemSevere dysuria, painful
hematuria, nocturia, pelvic pain, epithelial
inflammation similar to chronic interstitial cystitis, and unilateral or bilateral
hydronephrosis
Not investigated[73]
KetamineUnder drug effectsPattern of use: chronic misuse
Type: clinical study
Finality of use:
recreational
Dose and use
frequency: dose not informed, daily consume
Urinary systemDysuria, painful
hematuria, and post-voiding pain
Not investigated[74]
KetamineWithdrawal effectsPattern of use: chronic
Type: pre-clinical study
Finality of use: non-therapeutical
Dose and use
frequency: 100 or 300 mg/kg/day for 4 weeks
Urinary systemBladder inflammation,
interstitial nephritis, and
increased lymphocytes in bladder submucosal layer
Voiding dysfunction by neurogenic damage and dysregulation of
purinergic
transmission
[75]
KetamineUnder drug effectsPattern of use: acute
Type: pre-clinical study
Finality of use: not declared
Dose and use
frequency: 1000 µM/1x
Cardiovascular system (in vitro)Cerebral vasoconstrictionBlockade of Ca2+
activated K+ channels
[77]
KetamineUnder drug effectsPattern of use: acute
Type: pre-clinical study
Finality of use:
not declared
Dose and use
frequency: 1, 10, 100, 1000, and 10,000 µM/1x
Cardiovascular system (in vitro)Not specifiedInhibition of
sarcolemmal adenosine triphosphate-sensitive potassium (KATP)
channels mediated by SUR subunit with
specificity for
cardiovascular KATP channels
[78]
KetamineUnder drug effectsPattern of use: chronic
Type: pre-clinical study
Finality of use: abuse
Dose and use
frequency: 100 mg/kg/day for 4, 8, and 16 weeks
Urinary systemIncreased urination
frequency and decreased bladder capacity after 8 weeks
Increased
noncholinergic
contractions and P2X1 receptor expression in bladder
[80]
KetamineWithdrawal effectsPattern of use: chronic
Type: pre-clinical study
Finality of use: abuse
Dose and use
frequency: 30 mg/kg/day for 6 months
Urinary systemRelatively thinner bladder walls and infiltration of
mononuclear cells, similar to clinical situation of
interstitial cystitis
Decreased cholinergic neurons in urinary bladder by NMDA
receptor
overexpression
[81]
Ketamine plus
polydrugs
Under drug and
withdrawal effects
Pattern of use: chronic
Type: clinical study
Finality of use:
recreational
Dose and use
frequency:
not informed
Liver, biliary, and urinary systemCystitis and urinary
dysfunction; impaired liver function and biliary tree dilatation
Not investigated[49]
Ketamine plus
polydrug
Under drug effectsPattern of use: chronic
Type: clinical study
Finality of use:
recreational
Dose and use
frequency:
not informed
Liver, biliary,
cardiorespiratory, and urinary
system
Common bile duct
dilatation; increase in
serum levels of hepatic transaminases;
tachycardia; tachypnea and impaired respiratory function; overt hematuria associated with cramping abdominal pain, increased urinary frequency,
dysuria, moderate bilateral hydronephrosis, and cystitis
Not investigated[53]
Ketamine plus other drugs Pattern of use: chronic
Type: clinical study
Finality of use:
recreational
Dose and use
frequency:
not informed
Liver and biliary systemCommon bile duct
dilatation, microscopic bile duct injury, and liver
fibrosis
Not investigated[54]
Ketamine plus other drugsUnder drug effectsPattern of use:
not informed
Type: clinical study
Finality of use:
recreational
Dose and use
frequency:
not informed
Cardiovascular and urinary
system
Palpitations and chest pain, renal colic, urine leak, hematuria, and stranguriaNot investigated[36]
Ketamine plus other drugsUnder drug and
withdrawal effects
Pattern of use: chronic misuse
Type: clinical study
Finality of use:
recreational
Dose and use
frequency:
not informed
Respiratory
system
Authors report the effect of ketamine in producing impairment of pharyngeal and laryngeal reflexes,
diaphragm rigidity, and transient respiratory
depression
Not investigated[44]
Ketamine plus other drugsNot specifiedPattern of use: chronic misuse
Type: clinical study
Finality of use:
recreational
Dose and use
frequency: dose 0.125–5 g, frequency 3–7 days/week
Urinary systemPain in lower abdomen; burning during urination and urination
frequency increase;
incontinence and presence of blood in urine
Not investigated[38]
Ketamine plus ethanolUnder drug and
withdrawal effects
Pattern of use: chronic
Type: clinical study
Finality of use:
recreational
Dose and use
frequency:
not informed
Liver and biliary systemImpaired liver function and possible damage to bile ductsNot investigated[50]
Ketamine plus ethanolWithdrawal effectsLiterature reviewLiver, biliary, urinary, and cardiovascular systemRats: dysuria, increased collagen fibers in hepatic parenchyma, and tachycardia
Humans: signs and
symptoms of cystitis,
tachycardia, dysuria, and increase of liver fibrosis
Not investigated[61]
Ketamine plus ethanolWithdrawal and drug
effects
Pattern of use: chronic
Type: clinical study
Finality of use:
recreational
Dose and use
frequency: dose not informed;
occasional alcohol intake and daily ketamine inhalation
Liver and biliary systemIncrease in alkaline
phosphatase and
gamma-glutamyl
transpeptidase; concentric periductal fibrosis around bile ducts of varying sizes, consistent with primary or secondary sclerosing
cholangitis; interlobular bile ducts with thickened basement membranes, mild lymphocytic
infiltrates, and mild
ductular reaction
Not investigated[63]
EthanolUnder drug effectsLiterature reviewLiverLiver fibrosis and
cirrhosis, as a well as
synergic interaction to
infectious agents to induce liver injury
Production of
hepatotoxins,
activation of
redox-sensitive
transcription factors (i.e., nuclear factor kappa B (NF-κB),
neutrophils, and other immune cells’
recruitment), circulating pro-inflammatory
cytokines levels
increase, and intestinal dysbiosis with hepatic repercussions
[57]
EthanolUnder drug effectsLiterature reviewLiverLiver fibrosis and
cirrhosis, as a well as
synergic interaction to
infectious agents to induce liver injury
Production of
hepatotoxins, induction of oxidative stress,
activation of immune cells and circulating pro-inflammatory
cytokines levels
increase, deviation of
lipid metabolism
pathways, alteration of liver tissue remodeling factors activity, and
intestinal dysbiosis with hepatic
repercussions
[58]
EthanolUnder drug effectsLiterature reviewLiverLiver fibrosisAcetaldehyde leads to generation of reactive oxygen species and
activation of AP-1 and NF-kB transcription factors, resulting in pro-inflammatory
cytokines’ release and increased survival and remodeling activity of stellate liver cells
[59]
EthanolUnder drug effectsLiterature reviewCardiovascular systemAlcoholic cardiomyopathy and holiday heart
syndrome (chronic);
arrhythmias (chronic and acute); and acute atrial
fibrillation (acute)
Cardiovascular toxicity is a result of
dysregulation in
calcium and sodium conductance; beyond harmful effects on
cardiac contractile function, in general
after acute ethanol
exposure
[67]
EthanolUnder drug effectsLiterature reviewRespiratory
system
Chronic consumption
increases the risk of
developing lung diseases, such as acute lung injury, pneumonia, and
pulmonary fibrosis
Glutathione levels
decrease and TGF-1
expression increase; changes in tissue
remodeling and
extracellular matrix in lung tissue; activation of matrix
metalloproteinases (MMPs) in lungs,
particularly MMP-2 and MMP-9; and increase
in reactive oxygen
species generation
[71]
EthanolNot specifiedLiterature reviewUrinary systemLower urinary tract
symptoms
Urothelium
vulnerability to alcohol and its metabolites,
increased permeability, and lower urinary tract symptoms
[82]
EthanolWithdrawal effectsMeta-analysis of prospective chronic studiesUrinary systemKidney damage is
proportional to time of consumption and amount of alcohol, and there is a high prevalence of proteinuria in chronic users
Not investigated[83]
EthanolNot informedSystematic review and meta-analysisUrinary systemStatistically significant
inverse relationship of CKD in male adults with high alcohol consumption; and no significant
association between high alcohol consumption and risk of developing
proteinuria or end-stage renal disease
Not investigated[84]
EthanolWithdrawal effectsPattern of use: chronic
Type: pre-clinical study
Finality of use: abuse
Dose and use
frequency: ethanol 20% (v/v) daily for 24 weeks
Urinary systemIncrease of blood pressure, uric acid, and albumin;
and kidney changes
Activation of
renin–angiotensin system (RAS), oxidative stress and increased sympathetic nerve
activity
[85]
EthanolUnder drug effectsPattern of use: chronic
Type: pre-clinical study
Finality of use: abuse
Dose and use
frequency: ethanol 10% (v/v) daily for 12 weeks
Urinary systemReduction in creatinine clearance and urea in urine; and urea serum increaseNot investigated[86]
EthanolUnder drug effectsPattern of use: chronic
Type: pre-clinical study
Finality of use: abuse
Dose and use
frequency: ethanol 20% (v/v) daily for 12 weeks
Cardiovascular systemBlood pressure increaseIncreased aortic
inflammation; elevated angiotensin II levels;
induction of NADPH oxidase, leading to
endothelial injury; CuZn–SOD depletion; downregulation of
endothelial NO-generating system; and impaired vascular
vasodilatation in rats
[87]
EthanolWithdrawal effectsPattern of use: chronic
Type: pre-clinical study
Finality of use: abuse
Dose and use
frequency: ethanol 20% (v/v) daily for 6 weeks
Urinary systemIncrease in nitrite and
nitrate levels and tubular necrosis in proximal
tubules that is consistent in humans (acute tubular
necrosis)
Reactive oxidative
species generation and infiltration of
polymorphonuclear cells
[88]
EthanolUnder drug effectsPattern of use: chronic
Type: pre-clinical study
Finality of use: abuse
Dose and use
frequency: ethanol 2 g/kg daily for 10 and 30 weeks
Urinary systemKidney weight increase and new protein band with high molecular weight after 30 weeks of
ethanol treatment.
Reduction of reduced glutathione/oxidized glutathione, kidney
alcohol dehydrogenase activities increase, and distinct effects under antioxidant enzymes after 10 and 30 weeks
[89]
EthanolUnder drug effectsPattern of use: chronic
Type: pre-clinical study
Finality of use: abuse
Dose and use
frequency: ethanol 30% (v/v) daily for 2 months
Cardiovascular and urinary
system
Heart size augment and reduction of glomerular filtration rateIncrease of catalase
activity and decrease of lipid peroxidation in heart; opposite effects in kidney
[91]
Table 2. Effects reported in studies about ketamine and ethanol solely or combined on cognition, emotionality, and schizotypal parameters.
Table 2. Effects reported in studies about ketamine and ethanol solely or combined on cognition, emotionality, and schizotypal parameters.
Drug(s)Evaluation ConditionStudy InformationCNS Function/
Disorder
Main Effects
Described
Possible MechanismsReference
KetamineUnder drug and withdrawal
effects
Pattern of use: acute
Type: clinical study
Finality of use: abuse
Dose and use
frequency: 2 mg/kg daily
PsychosisImpaired on semantic memory tasks; higher levels of dissociation and schizotypal
symptoms
Not investigated[24]
KetamineWithdrawal
effects
Pattern of use: chronic misuse
Type: clinical study
Finality of use: abuse
Dose and use
frequency: dose not
informed; frequency— more than 4 times a week.
Cognition and
depression
Short- and long-term memory impairments, vulnerability to
dangers behavior, and depressive symptoms
Not investigated[40]
KetamineWithdrawal
effects
Pattern of use: chronic
Type: clinical study
Finality of use: abuse
Dose and use
frequency: dose not
informed; frequent use
Cognition and
depression.
Frequent use: reduced psychological
well-being and broad range of cognitive
impairments.
Infrequent use:
increased symptoms of thought disorder,
delusions, and
dissociation, but not cognitive impairment
Not investigated[39]
KetamineUnder drug
effects
Pattern of use: acute
Type: clinical study
Finality of use:
not specified
Dose and use
frequency: single
administration of 0.4 mg/kg and 0.8 mg/kg.
PsychosisIncreased
schizophrenic and
dissociative symptoms after ketamine use; psychotomimetic
effects of ketamine are detectable on clinical scales
Not investigated[103]
KetamineUnder drug
effects
Pattern of use: acute
Type: clinical study
Finality of use: clinical
Dose and use
frequency: 0.5 mg/kg
infused over 40 min
EmotionalitySubacute increase of prefrontal connectivity associated to
antidepressant
response
Ketamine seems activate prefrontal glutamate neurotransmission,
contributing to transient
psychotomimetic effects and delayed and sustained antidepressant
effects
[114]
KetamineUnder drug
effects
Pattern of use: acute
Type: clinical study
Finality of use:
subanesthetic
Dose and use
frequency: 0.3 mg/kg for 2 weeks
PsychosisIncrease in schizotypal symptomsFlow activation on anterior
cingulate and prefrontal cortex;
decreased flow activation on visual cortex and hippocampus; and
abnormal glutamatergic
transmission involved on
pathophysiology of psychotic symptoms
[117]
KetamineNot specifiedLiterature reviewCognition and psychosisCognitive impairment; psychotic and negative symptomsReduction of NMDA receptors
related to negative symptoms
[108]
KetamineUnder drug
effects
Pattern of use: acute
Type: clinical study
Finality of use:
non-therapeutical
Dose and use
frequency: 15 mg/kg for 5 min to induce
psychopathological
effects, followed by dose of 0.014 mg/kg/min for 90 min
Emotionality and psychosisAffect changes,
illusions,
hallucinations, and ego dissolution
Inhibition of NMDA receptors to increase in DA levels in striatum, inducing euphoria- and
mania-related features
[119]
KetamineUnder drug and withdrawal
effects
Pattern of use: acute
Type: pre-clinical study
Finality of use:
not informed
Dose and use
frequency:
2, 10, and 20 mg/kg (i.p(i.p.), or once, or twice; or 25 mg/kg for 7 days
CognitionHighest ketamine dose elevates levels of errors of omission in
attentional tasks, whereas disrupted
attentional performance during
pre-treatment period
Increase in cortical acetylcholine
release
[121]
KetamineUnder drug
effects
Pattern of use: acute
Type: pre-clinical study
Finality of use:
subanesthetic
Dose and use
frequency: single dose 150 mg/kg
Schizotypal
behaviors
Suppression of
high-frequency EEG activity and disruption of cortical coherence
Increased concentrations of cortical ACh in active arousal systems in setting of unconscious state[122]
KetamineUnder drug
effects
Pattern of use:
not informed
Type: pre-clinical study
Finality of use:
not informed
Dose and use
frequency: a single administration of 0.1 nM to 10,000 nM
Psychosis (in vitro) Biological events
related to
schizophrenia
Ketamine binds to D2 receptors, increasing dopamine
neuro-availability; and antagonism of NMDA receptors induces
schizotypy
[123]
KetamineUnder drug
effects
Pattern of use: acute
Type: pre-clinical study
Finality of use:
not specified
Dose and use
frequency: a single administration of 0.01 a 100 µM
Psychosis (in vitro) Partial agonism on D2, and 5-HT2 (on a smaller scale) receptorsKetamine binds to D2 receptors,
increasing the neuro-availability of dopamine; small affinity to 5-HT2 receptors; and other mechanisms induce non-selective multi-system neurochemical perturbation
[124]
KetamineUnder drug
effects
Pattern of use: acute
Type: pre-clinical study
Finality of use:
therapeutical
Dose and use
frequency: 30 mg/kg and local administration of 0.1 mM
EmotionalityIncreased serotonin
release in medial
prefrontal cortex
Increase of serotonin release on
medial prefrontal cortex through cholinergic neurons projected
pedunculopontine tegmental
nucleus to dorsal raphe nucleus
[125]
KetamineNot specifiedLiterature reviewPsychosisSchizotypal symptomsGlutamatergic hypoactivity through antagonism of NMDA receptors may be associated with
schizophrenia manifestation
[126]
KetamineUnder drug
effects
Literature reviewCognitionAbusive use of
ketamine induces
cognitive damage
NMDA receptors blockade on gamma-aminobutyric acid (GABA) neurons on thalamic reticular
nucleus leads to disinhibition of
dopaminergic neurons and
increased dopamine release
[142]
KetamineWithdrawal
effects
Pattern of use: acute
Type: pre-clinical study
Finality of use: abuse
Dose and use frequency: 10 mg/kg for 3 days
Cognition and emotionalityMemory impairment, anxiogenic and
depressive behavior
Oxidative stress in hippocampus[25]
KetamineWithdrawal
effects
Pattern of use: chronic
Type: clinical study
Finality of use: abuse
Dose and use frequency: 3.4 g/day; frequency varied between 1 time per day, more than 4 times per week, and less than 4 times per week
Cognition and emotionalityDepressive,
anxiogenic, and
psychotic symptoms
Not investigated[165]
KetamineUnder drug and withdrawal
effects
Pattern of use: acute
Type: pre-clinical study
Finality of use:
anesthetic and
subanesthetic
Dose and use
frequency: 2 mg/kg daily for 2 days
AnxietyDoses employed have no effects on anxiety- or panic-related
behaviors
Not investigated[167]
KetamineWithdrawal
effects
Pattern of use: chronic
Type: clinical study
Finality of use: abuse
Dose and use
frequency: not informed
Dissociative symptomsGreater affective symptoms and
perceptual
disturbances
Not investigated[93]
KetamineWithdrawal
effects
Pattern of use: chronic
Type: clinical study
Finality of use: abuse
Dose and use
frequency: not informed
Cognition,
emotionality and schizotypal
symptoms
Semantic memory
deficiencies decrease but are reversible with marked reduction in use; impairment in
episodic memory and attentional
functioning;
schizotypal symptoms and perceptual
distortions may persist after discontinuing ketamine use
Not investigated[104]
KetamineWithdrawal
effects
Pattern of use: acute
Type: clinical study (case report)
Finality of use:
anesthetic
Dose and use frequency 650 mg/kg to 15 mg/kg for 7 days
General
behavioral
alterations
Fever, sleep inversion, restlessness, and
drooling (24 and 48 h after withdrawal); 17 days later, behavior and cognitive
impairment, including aggression and
language deficits, in addition to affecting motor skills
Not investigated[175]
KetamineWithdrawal
effects
Pattern of use: acute
Type: pre-clinical study
Finality of use:
anesthetic
Dose and use
frequency: single dose of 20 mg/kg
subcutaneously
CognitionImpairment in
short-term recognition memory
Decreased cellular viability in
hippocampus, and long-term increase in hippocampal glutamate uptake. Prevention of vulnerability to
glutamate-induced neurotoxicity in frontal cortex of adult rats
[174]
KetamineUnder drug
effects
Pattern of use: chronic
Type: clinical study
Finality of use: abuse
Dose and use
frequency: at least an average use of one vial per week or more over the last 3 months
MemoryWorking memory not affectedDorsolateral prefrontal cortex
D1 receptor upregulation; binding potential upregulation
significantly correlated to number of vials of ketamine used per week.
[181]
EthanolWithdrawal
effects
Pattern of use: chronic
Type: clinical study
Finality of use: abuse
Dose and use
frequency: not informed
PsychosisHallucinations of
schizophrenic origin and hallucinations from alcohol are very similar
Not investigated[132]
EthanolWithdrawal
effects
Pattern of use: chronic
Type: clinical study
Finality of use: abuse
Dose and use
frequency: 200 g/day for 30 years
General
behavioral
alterations
Finger tremor, mildly altered liver enzymes, and decreased regional cerebral blood flowNot investigated[136]
EthanolWithdrawal
effects
Pattern of use: chronic
Type: clinical study
Finality of use: abuse
Dose and use
frequency: 400 g/day of alcohol for a long time (undefined)
Psychotic stateHallucinationsDamage to thalamic structures
associated with manifestation of
hallucinations
[137]
EthanolWithdrawal
effects
Pattern of use: chronic
Type: clinical study
Finality of use: abuse
Dose and use
frequency: unreported dose and consume
frequency at least 10 years
Psychotic stateVerbal hallucinations, hallucinatory
delusions, and
affective frustration (mainly alarm and fear) in a state of clear consciousness
Acute alcoholic hallucinosis is linked to changes in excitatory and inhibitory transmission in the brain[138]
EthanolWithdrawal
effects
Pattern of use: acute
Type: clinical study
Finality of use: abuse
Dose and use
frequency: unreported dose; ex-user patients undergoing treatment
Psychotic stateChronic alcohol
consumption induces hallucinogenic events, and it alters plasmatic
concentration of
tyrosine, tryptophan, and phenylalanine
Amino acid imbalances result in
decreased brain serotonin levels and increased brain dopamine,
inducing hallucinatory experiences
[139]
EthanolUnder drug or withdrawal
effects
Pattern of use: chronic
Type: pre-clinical study
Finality of use: abuse
Dose and use
frequency: 2 g/kg/day or 10%; daily frequency
Rewarding
system
Broader effects on
dopaminergic system in voluntary ethanol intake model
Increased dopamine degradation, influencing the reward system[140]
EthanolUnder drug and withdrawal
effects
Literature reviewGeneral
behavioral
alterations
Withdrawal symptoms, delirium tremens,
Wernicke–Korsakoff syndrome, and fetal
alcohol syndrome
Acute effects of ethanol disrupt
glutamatergic neurotransmission by NMDA blockade. Prolonged
inhibition of NMDA receptor
results in development of
supersensitivity. Acute absence of ethanol increases postsynaptic
neurons activity (i.e., noradrenergic system and glutamate-induced
excitotoxicity)
[143]
EthanolUnder drug
effects
Literature reviewReward systemBinge drinking induces decreased reward
neurocircuitry function and recruitment of
anti-reward/stress mechanisms
Ethanol interacts with NMDA, GABAA, glycine, 5-HT, and
nicotinic receptors, as well as
L-type Ca2+ channels and G protein-activated internal rectifier K+ channels of neurotransmitters/neuropeptides leading to typical acute behavioral effects of alcohol
[144]
EthanolWithdrawal
effects
Literature reviewGeneral behavioral alterationsAnxiety, depression, tremors, rigidity,
hyperactivity,
convulsion, coma, and even death
Intense generation of reactive
oxygen species and activation of stress-responding protein kinases
[145]
EthanolUnder drug and withdrawal
effects
Literature reviewCNS adaptationsBrain excitotoxicityHyperexcitability after alcohol withdrawal may contribute to
excitotoxicity. Alterations in
function and/or expression of
glutamate, GABA, and
voltage-activated calcium channels contribute to hyperexcitability
[153]
EthanolUnder drug and withdrawal
effects
Literature reviewGeneral behavioral alterationsDepressive episodes, severe anxiety,
insomnia, suicide, and abuse of other drugs
Not investigated[154]
EthanolUnder drug and withdrawal
effects
Literature reviewCNS impairmentsAcute: excitement, ataxia, and lethargy. Chronic: cognitive, emotional, and motor disturbances.
Withdrawal: autonomic
hyperactivity, tremor, anxiety,
restlessness, seizures, hallucinations, and
delirium
Ethanol stimulates microglia,
inducing neuroinflammation that triggers neuropathogenic processes
[155]
EthanolWithdrawal
effects
Pattern of use: chronic
Type: clinical study
Finality of use: abuse
Dose and use
frequency: unreported dose and frequency
Cognition and emotionalityDepressive disorders, anxiety, sleep
disturbance,
impairment of
cognitive ability, and increased plasma
macrophage-derived chemokine
Macrophage-derived chemokine is associated with alcoholism, phobia, and interpersonal sensitivity[156]
EthanolWithdrawal
effects
Pattern of use: chronic
Type: pre-clinical study
Finality of use: abuse
Dose and use
frequency: 4% v.o. for 26 months
AnxietySocial isolation and
increased cortisol
Not investigated[171]
EthanolWithdrawal
effects
Pattern of use: acute
Type: pre-clinical study
Finality of use: abuse
Dose and use
frequency: vapor
exposure for 7 days; blood alcohol levels 127 mg%
Endocrine
regulation
Not specifiedHypothalamic–pituitary adrenal axis activity alterations[172]
EthanolUnder drug
effects
Pattern of use: acute
Type: clinical study
Finality of use: abuse
Dose and use
frequency: a single dose of 0.8 g/kg
Cognition and emotionalityNegative effects on
several dimensions of mood; impaired verbal memory; and dysphoria
Norepinephrine mediates
behavioral alterations ethanol-induced (i.e., dysphoria), while
serotonin provokes opposite
activity
[158]
EthanolUnder drug
effects
Pattern of use: acute
Type: pre-clinical study
Finality of use: abuse
Dose and use
frequency: a single administration of 3.5 g/kg (i.p.)
CNS monoamine dependent
functions
Not investigatedMesolimbic and nigrostriatal
dopaminergic systems’
hyperactivation, increasing DA
release and catabolism
[159]
EthanolUnder drug
effects
Pattern of use: acute
Type: pre-clinical study
Finality of use: abuse
Dose and use
frequency: 1, 2, 3, and 4 g/kg for 7 days
CNS monoamine dependent
functions
Not investigatedDopamine, NE, and metabolite’s levels decrease on dorsal raphe and Locus coeruleus[160]
EthanolNot informedLiterature reviewGeneral
behavioral
alterations
Psychomotor
depression, difficulties in information storage and logical reasoning, motor incoordination, stimulation of reward system
Direct action on GABA, glutamate, and endocannabinoids systems;
indirect action on limbic and opioid system; action on calcium channels, potent and proteins regulated by GABA hippocampus, in addition to central actions not mediated by
vitamin B1 deficiency
[161]
EthanolUnder drug and withdrawal
effects
Literature reviewCognitionNutritional disorders and dementiaLoss of hippocampal CA1 and CA3 pyramidal neurons, mossy
fiber-CA3 synapses and dentate granule cells, and cholinergic
neurons in basal forebrain;
pathological neuroadaptations (i.e., excitatory/inhibitory neurotransmitters balance and oxidative stress)
[182]
Ketamine plus ethanolUnder drug
effects
Pattern of use:
sub-chronic
Type: pre-clinical study
Finality of use: abuse
Dose and use
frequency: 2 or 4 g/kg orally of ethanol plus 30 mg/kg i.p. ketamine for 14 days
SchizotypyIncreased activity,
stereotyped behavior, ataxia, and
morphological changes, as well as severe neurotoxicity
Altered behaviors were associated with alcohol-induced increases in ketamine-induced higher levels of Glu and DA in cortex and hippocampus[141]
Ketamine plus ethanolUnder drug
effects
Pattern of use: acute
Type: pre-clinical study
Finality of use: abuse
Dose and use
frequency: 10% ethanol solutions plus 0.28 mg/mL ketamine for 35 days, for 1 h
AnxietyEthanol:
anxiogenic-like
behavior.
Ketamine:
anxiolytic-like
behavior.
Ketamine + ethanol:
absence of effect
Ketamine interfered in development of tolerance to anxiolytic
effects of ethanol through
modulation of several subsystems targeted by both drugs
[173]
Ethanol plus other drugs Pattern of use: chronic
Type: pre-clinical study
Finality of use: abuse
Dose and use
frequency: 3 g/kg/day, 3 times a week for 5 weeks
Cognition and emotionalityCognitive impairment; depressive and
anxiety-like behavior
Alteration in peripheral markers of oxidative stress: decrease in nitrite levels; increase of sulfhydryl groups, MDA levels, superoxide dismutase, and catalase activity[157]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kobayashi, N.H.C.; Farias, S.V.; Luz, D.A.; Machado-Ferraro, K.M.; Conceição, B.C.d.; Silveira, C.C.M.d.; Fernandes, L.M.P.; Cartágenes, S.d.C.; Ferreira, V.M.M.; Fontes-Júnior, E.A.; et al. Ketamine plus Alcohol: What We Know and What We Can Expect about This. Int. J. Mol. Sci. 2022, 23, 7800. https://doi.org/10.3390/ijms23147800

AMA Style

Kobayashi NHC, Farias SV, Luz DA, Machado-Ferraro KM, Conceição BCd, Silveira CCMd, Fernandes LMP, Cartágenes SdC, Ferreira VMM, Fontes-Júnior EA, et al. Ketamine plus Alcohol: What We Know and What We Can Expect about This. International Journal of Molecular Sciences. 2022; 23(14):7800. https://doi.org/10.3390/ijms23147800

Chicago/Turabian Style

Kobayashi, Natalia Harumi Correa, Sarah Viana Farias, Diandra Araújo Luz, Kissila Márvia Machado-Ferraro, Brenda Costa da Conceição, Cinthia Cristina Menezes da Silveira, Luanna Melo Pereira Fernandes, Sabrina de Carvalho Cartágenes, Vânia Maria Moraes Ferreira, Enéas Andrade Fontes-Júnior, and et al. 2022. "Ketamine plus Alcohol: What We Know and What We Can Expect about This" International Journal of Molecular Sciences 23, no. 14: 7800. https://doi.org/10.3390/ijms23147800

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop