Next Article in Journal
Adapting a Dementia Care Management Intervention for Regional Implementation: A Theory-Based Participatory Barrier Analysis
Previous Article in Journal
Association of Esophageal Cancer Mortality with Municipal Socioeconomic Deprivation Level in Japan, 2013–2017: An Ecological Study Using Nationwide Data
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Commentary

Proposing a “Brain Health Checkup (BHC)” as a Global Potential “Standard of Care” to Overcome Reward Dysregulation in Primary Care Medicine: Coupling Genetic Risk Testing and Induction of “Dopamine Homeostasis”

1
The Kenneth Blum Institute on Behavior & Neurogenetics, Austin, TX 78701, USA
2
Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
3
Department of Psychiatry, Tulane School of Medicine, New Orleans, LA 70112, USA
4
Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel
5
Future Biologics, Lawrenceville, GA 30043, USA
6
Division of Addiction Research & Education, Center for Psychiatry, Medicine & Primary Care (Office of Provost), Western University Health Sciences, Pomona, CA 91766, USA
7
Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA
8
The Molecular Neuropsychiatry Research Branch, NIH National Institute on Drug Abuse, Baltimore, MD 21224, USA
*
Author to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2022, 19(9), 5480; https://doi.org/10.3390/ijerph19095480
Submission received: 17 February 2022 / Revised: 26 April 2022 / Accepted: 28 April 2022 / Published: 30 April 2022

Abstract

:
In 2021, over 100,000 people died prematurely from opioid overdoses. Neuropsychiatric and cognitive impairments are underreported comorbidities of reward dysregulation due to genetic antecedents and epigenetic insults. Recent genome-wide association studies involving millions of subjects revealed frequent comorbidity with substance use disorder (SUD) in a sizeable meta-analysis of depression. It found significant associations with the expression of NEGR1 in the hypothalamus and DRD2 in the nucleus accumbens, among others. However, despite the rise in SUD and neuropsychiatric illness, there are currently no standard objective brain assessments being performed on a routine basis. The rationale for encouraging a standard objective Brain Health Check (BHC) is to have extensive data available to treat clinical syndromes in psychiatric patients. The BHC would consist of a group of reliable, accurate, cost-effective, objective assessments involving the following domains: Memory, Attention, Neuropsychiatry, and Neurological Imaging. Utilizing primarily PUBMED, over 36 years of virtually all the computerized and written-based assessments of Memory, Attention, Psychiatric, and Neurological imaging were reviewed, and the following assessments are recommended for use in the BHC: Central Nervous System Vital Signs (Memory), Test of Variables of Attention (Attention), Millon Clinical Multiaxial Inventory III (Neuropsychiatric), and Quantitative Electroencephalogram/P300/Evoked Potential (Neurological Imaging). Finally, we suggest continuing research into incorporating a new standard BHC coupled with qEEG/P300/Evoked Potentials and genetically guided precision induction of “dopamine homeostasis” to diagnose and treat reward dysregulation to prevent the consequences of dopamine dysregulation from being epigenetically passed on to generations of our children.

1. SUD Global Pandemic

Addiction is a global pandemic that has negatively affected millions of people [1]. Although addiction itself is not a diagnostic term in the International Classification of Diseases 10th revision (ICD-10), it is still widely used by professionals and the public [1]. When it comes to diagnostic terminology, “addiction” was previously classified as substance abuse and substance dependence in the fourth edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM) [2]. The DSM-5 consolidated the terminology into substance use disorder (SUD) with a severity scale [3]. However, federal agencies continue to use different/multiple terms such as SUD, alcohol use disorder (AUD), opioid use disorder (OUD), addiction, substance dependence, substance abuse, and experimental gateway, etc. [4,5,6,7,8]. For the purpose of this paper, SUD is all-encompassing.
The National Center for Drug Abuse Statistics (NCDAS) reports that ~12% of Americans over the age of 12 use illicit drugs, and if alcohol and tobacco are included, then the percentage increases to ~60 [9]. In the United States (US), ~50% of teenagers have misused a drug at least once and ~61% of teenagers have abused alcohol by 12th grade [10,11]. In addition, 70% of users who experiment with illegal drugs before age 13 develop SUD within the next 7 years, compared to 27% of those who experiment with illegal drugs after age 17 [9]. Furthermore, many individuals with SUD also suffer from polysubstance abuse. For example, according to the Substance Abuse and Mental Health Services Administration (SAMHSA), ~35% of adults with illicit drug use disorder also have AUD [12]. Alcohol is one of the most commonly used drugs, and AUD is the most common type of SUD in the US [13,14]. The lifetime prevalence of AUD is ~29%, the prevalence of binge drinking in young adults (age 18–25) is ~31%, and ~70% of Americans report having alcohol in the past year [9,10,15]. SUD global severity, by country, is also underreported and inconsistent, ranging from 5 to 20%, depending on the agency [16,17,18,19,20,21,22].
SUD reporting is underestimated in the US due to stigma, confusion, lack of precision in reporting medical causes of death, poor self-reporting, and confusing/lack of uniform terminology, etc. [23,24,25,26,27,28,29,30,31,32,33,34,35]. Patients with SUD often have co-occurring neuropsychiatric/medical disorders, and thus, their deaths can be attributed to any one of their comorbidities, leading to death certificate discrepancies. Furthermore, SUD is often the tipping point to death in patients with medical comorbidities [36]. Drug overdose deaths are often reported as accidents (cars, machinery, military, prescription mistakes, etc.), heart attacks, sudden cardiac deaths, strokes, etc. [23,24,25,26]. In 2019, SUD was reported as the third and/or tenth leading cause of death classified as accidental or suicide, respectively [37]. Approximately 50% of all death certificates and 20–30% of drug overdose death certificates that the CDC receives are inaccurate or have incomplete causes of death [25,35]. These inaccuracies result in the underreporting of deaths attributed to SUD. SUD mortality will likely continue to increase until a more accurate and reliable system is implemented [35]. Finally, we believe that in those cases with the presence of DNA antecedent polymorphisms across the brain reward circuitry, it might suggest that SUD is not easily reversed and may indeed be a life-long condition if untreated. Nevertheless, our new knowledge related to the role of environment and epigenetic influence on gene expression that includes acetylation (positive expression) and methylation (negative expression), the presence of SUD in an individual could become a reversable phenotype if the epigenetic repair becomes a life-style change, including induction of brain circuitry homeostasis.
Therefore, we believe that the US medical system, especially pediatrics, must implement and utilize standard neuropsychiatric metrics and imaging in the diagnosis and treatment of SUD until the number of SUD deaths becomes zero.

2. Economic Burden of SUD Crisis

During the COVID-19 pandemic, reported opioid youth deaths increased by 25%, from ~75,000 to ~100,000, with an average age of 24, ~70% men [38,39,40,41]. SUD is estimated to cost the US ~1 trillion dollars, i.e., 700 billion National Institute on Drug Abuse (NIDA), 300 billion National Institute on Alcohol Abuse and Alcoholism (NIAAA), but this value is extremely underestimated [42,43,44,45,46,47,48,49,50]. If we include all SUD, + food, + neuropsychiatric/medical comorbidities, the annual cost might be as high as 4 trillion, including lost work productivity [36,42,43,44,45,46,47,48,49,50,51]. Furthermore, a recent study involving young individuals (age 10–24) showed that, over a 5-year period (2015–2019), ~1.25 million years of life were lost [52]. Even with current epidemiological data, the loss to the US is approximately 7 million dollars in life years (loss of approximately 50 years of a person’s life by dying at 24 of SUD) [45,47]. If an average person’s lifetime financial productivity is ~USD 4 million dollars, then the loss to the gross national product (GNP)/future economy is ~USD 30 trillion dollars over time [45,47]. The direct and indirect medical comorbidities of SUD are so vast due to reward deficiency syndrome that it may account for up to 75% of the total health expenditure, or ~USD 4 trillion dollars (Table 1) [36,42,43,44,45,46,47,48,49,50,51,53].
Secondary costs of SUD, especially food addiction/obesity, are in the trillions of dollars because of medical, neuropsychiatric, and social comorbidities (Table 1 and Table 2) [36,42,43,44,45,46,47,48,49,50,51,168]. Most SUD patients also concomitantly suffer from an abundant number of neuropsychiatric and medical findings, referred to by some as global dopamine dysfunction. In contrast, the premorbid neuropsychiatric medical findings that predict progression to SUD are less well known (Table 2). However, some common precursors to SUD include school failure and problems with family, memory, and attention [32,33,34] (Table 2).

3. Failure of the Current Paradigm

NIDA defines SUD as a chronic relapsing disorder. As a result, the long-term success rate of Alcoholics Anonymous (AA) and rehab groups is very low, except on the rare occasion when patients follow treatment guidelines [6,181,182]. According to NIDA, the relapse rate for SUD is 40–60% and “resembles those of other chronic diseases such as diabetes, hypertension, and asthma” [183]. Therefore, longitudinal studies must follow patients with SUD over the course of years (we recommend at least five) or even a lifetime for verified results [184,185]. Furthermore, a placebo response may last as long as a year with the buoyant attention of a healthcare practitioner [186].
Neuropsychiatric disorders are common in the US, and according to the National Institute of Mental Health, nearly one in five US adults has a mental illness [187]. In addition, ~45% of individuals with SUD have co-occurring neuropsychiatric disorders [12]. Neuropsychiatric disorders and SUD influence each other, and their combined presentation can result in more severe functional impairment, poorer treatment outcomes, higher treatment costs, and increased mortality and morbidity [188]. Many individuals with SUD and/or neuropsychiatric disorders do not receive treatment [187,188]. In a study by Han et al., only ~9% of adults with SUD and co-occurring neuropsychiatric disorders received both mental health care and substance use treatment, while ~52% received neither form of treatment [188]. Additionally, they reported that ~3–13% received only substance use treatment, and ~34–44% received only mental health care [188]. These findings reveal a significant disparity between the prevalence of SUD and co-occurring neuropsychiatric disorders prevalence and treatment rates.
Solving the SUD pandemic would resolve the US economic crisis, including debt service. One important dilemma related to the treatment of opioid dependence is the widespread use of opioids to specifically treat the unfortunate victims of OUD. While the prescribing of opioids (i.e., Buprenorphine) has reduced harm, it does not result in prophylaxis [189]. To achieve some reasonable solution in terms of treatment, relapse prevention, and prophylaxis, one novel therapeutic should include genetic risk assessment and induction of dopamine balance [190]. Many addiction programs are opting for the use of the non-addicting and relatively safe narcotic antagonist Naltrexone despite its associated poor compliance, even in the injectable form [191,192,193]. An analysis conducted at Stanford University suggested that the number of unwanted deaths from prescription opioids and street heroin will continue to increase if no changes are made to the currently available treatment, prevention, and public health approaches [194].
Studies have shown that Naltrexone is beneficial by attenuating cravings via “psychological extinction” and reducing relapse. In addition, research performed by Blum’s group has shown that Buprenorphine is the current medication-assisted treatment (MAT) of choice, but injectable Naltrexone plus an agent to enhance dopaminergic function and tone may rekindle interest amongst addiction physicians and patients [195]. Previously, an open-label investigation in humans showed improvement in Naltrexone compliance and outcomes with dopamine augmentation using the pro-dopamine regulator referred to as KB220 (262 days) compared to naltrexone alone (37 days) [195]. This well-studied complex consists of amino-acid neurotransmitter precursors and enkephalinase inhibitor therapy compared to standard treatment [196]. Consideration of this novel paradigm shift may assist in not only addressing the current opioid epidemic but also the broader issue of reward dysregulation.
It is important to recognize that SUD may also occur with certain palatable foods such as sugar and may also become highly addictive to the individual. It is well known that over 90 percent of people with obesity have an abnormal romance with carbohydrates. It is also well-known that there is strong evidence that common neurochemical mechanisms are observed in both animal and human imaging studies [48,180].

4. SUD—A Neurological Disorder

SUD is widely recognized as a neurological disorder. However, despite this, there are currently no standard, routine, objective brain assessments being performed, and without standardization, patients with neurological/brain disorders/dysfunction will not be able to receive the treatment they need. For example, it took approximately 40 years to obtain a core set of cardiac tests, e.g., electrocardiogram (EKG), echocardiogram, and blood tests, etc., which ultimately halted the bypass cardiac pandemic. Brain health needs to have a similar stepwise approach that parallels those used to treat cardiac disease because, without routine objective assessments, clinical studies lack reliability, accuracy, dependability, and reproducibility (Table 3). Therefore, utilizing the currently available inexpensive objective testing of premorbid memory, attention, and neuropsychiatry, with the possible addition of supplemental imaging and genotyping, would greatly improve brain/mind health and help combat the addiction pandemic (Table 3) [180,197].

5. Our Theoretical Construct for Primary Care and Reward Dysregulation

Based on the literature review, it is our opinion that a novel, cost-effective Brain Health Checkup that involves the domains: Memory, Attention, Neuropsychiatry, and Neurological Imaging may become the new standard of care in pediatric medicine, starting with children aged five and especially following Genetic Addiction Risk Score (GARS) testing (for curiosity or especially in a family tree robust with Reward Deficiency Syndrome (RDS) behaviors such as AUD, etc.). This checkup would be referred to as a “Brain Health Checkup” (BHC). Utilizing primarily PUBMED, the current paper herein, reviews over 36 years of virtually all the computerized and written-based assessments of Memory, Attention, Neuropsychiatry, and Neurological imaging. This research found the following tests to be the most beneficial for each of the aforementioned domains and recommends their use in the BHC: Central Nervous System Vital Signs (Memory), Test of Variables of Attention (Attention), Millon Clinical Multiaxial Inventory III (Neuropsychiatric), Quantitative Electroencephalogram/P300/Evoked Potential (Neurological Imaging). Obviously, these denoted domains should not be limited but in our opinion be included in any BHC to be effective in diagnoses in primary care and reward dysregulation.

6. SUD Induced Memory Loss

SUD is a total brain disorder that causes brain injury, delayed processing, decreased memory, attention abnormalities, neuropsychiatric disturbance, and MRI results that parallel dementia/mild cognitive impairment (MCI) (Table 4) [197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226]. Brain atrophy/damage is found in MCI, moderate cognitive impairment (MoCI), and dementia [197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226]. MRI results can be confusing if not performed serially, as the worsening of atrophy is more predictive of dementia. SUD may cause more injury to the dorsal lateral prefrontal cortex and mesial temporal lobe [198]. Additionally, studies have shown that SUD may cause slightly greater damage than MCI [197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226]. The mechanism of SUD brain atrophy is the same as dementia, called retrogenesis, which is the opposite of neurogenesis [198,227,228,229,230,231,232,233,234,235].
Out of all the memory tests reviewed, the Central Nervous System Vital Signs (CNSVS) assessment was found to be the best. The CNSVS is a computerized neuropsychological test used to evaluate the neurocognitive status of patients, which includes mental processes that range from simple motor performance, attention, and memory to executive functions [236,237]. It includes 10 normed objective neurocognitive tests: verbal memory, visual memory, finger tapping, symbol digit coding, shifting attention, continuous performance, perception of emotions, Stroop test, non-verbal reasoning, and four-part continuous performance [236,237]. CNSVS was found to be the best memory test because it is SOC2 certified, registered with the FDA, available globally, economical at ~USD 35, has greater than 99% compliance, and only takes ~30 min to complete [236,237,238,239,240]. CNSVS has high appeal because complex attention correlates with better SUD function and possible prevention [236,238,239,241,242]. CNSVS has no practice learning effect (test–retest probability) and becomes a reliable indicator of deterioration or improvement over time [236]. The presidential Montreal Cognitive Assessment (MoCA) test correlates well with CNSVS results [242].

7. SUD Induced Attention Issues

Attention deficits are common in SUD patients [243,244,245,246]. In fact, research has shown that of adults presenting with SUD, 20–30% have concurrent ADHD, and 20–40% of adults diagnosed with ADHD also have a history of SUD [180,243,244,245,246]. Therefore, the Test of Variables of Attention (TOVA), which is an attention screening test, would be useful in the diagnosis and subsequent treatment of SUD. The TOVA measures omission errors (inattention), commission errors (impulsivity), response time, response time variability (consistency), commission error response time, post-commission error response time, anticipatory responses, embedded performance validity, inattentiveness, impulsivity, sustained attention, and vigilance [247,248]. It can successfully diagnose ADHD subtypes, including inattention, impulsivity, attentional failure due to depression or psychomotor retardation, and inconsistency [248,249]. TOVA is currently the best attention test because it is FDA approved, economical at ~USD 15, has greater than 99% compliance, and only takes ~22 min to complete [249]. In addition, TOVA does not have a significant practice effect, is easy to supplement with Conners’ checklist, and allows for diagnostic heterogeneity [247,249,250].

8. Neuropsychiatric/Global Dopamine Dysfunction

Virtually all SUD patients have some form of neuropsychiatric/global dopamine dysfunction in life. Premorbid neuropsychiatric medical findings that predict progression to SUD are not widely screened for or even known (Table 2). The range of neuropsychiatric consequences of SUD are extremely diverse and cannot be accurately or reliably measured without standardization of testing (Table 5). This results in the absence of early identification of high-risk individuals across primary care and the various specialties. If a BHC were a mandated part of neuropsychiatric medicine, then the premorbid states would be characterized. For example, cardiovascular disease has been greatly controlled by testing premorbid patients with no heart disease or only early disease (hypertension or hyperlipidemia), i.e., EKG, labs, and echocardiogram. This approach engages the patient in their treatment and has been repeated in diabetes (hemoglobin A1c), obesity (body mass index, bioimpedance), breast cancer (mammogram/ultrasound), cervical cancer (pap smear), asthma (pulmonary function tests), and routine blood work for anemia, kidney, liver disease, etc. In addition, the BHC would help destigmatize SUD brain disorders, which is a key step towards medical parity [30,31]. The individual brain function tests could also be called BFTs to parallel liver function tests (LFTs), thyroid function tests (TFTs), pulmonary function tests (PFTs), and kidney function tests (KFTs), etc. [30,31].
The Millon Clinical Multiaxial Inventory-III (MCMI-III) is a diagnostic tool that can quickly and efficiently screen patients for neuropsychiatric disorders (Table 6). The MCMI-III provides a measure of 24 Axis I and II disorders and clinical syndromes for adults undergoing psychological or psychiatric assessment/treatment [252,253]. It has 175 questions and 27 scales with 99% compliance [252,253]. Assessed MCMI 1–4 correlates with the DSM [252,253,254,255]. MCMI-III contains a lifetime interpretation guide that captures many past and future predictions of behavior [256]. Non-psychiatrists should ideally use MCMI-III because DSM 5 has over 500 diagnoses/severity, which makes it difficult for non-psychiatrists to use. Additionally, psychiatrists’ use of the DSM typically results in one to two diagnoses for SUD patients, which is often inaccurate and misleading because SUD patients are typically worse than any one or two diagnoses alone. The most common finding of the MCMI-III test is the rate of depression, ~10–20% [257]. We studied the impact of SUD on brain electrophysiology and found that SUD significantly worsens normal temporal lobe abnormalities [257]. The bitemporal injury of SUD increases mood disorders [257]. Our MCMI-III data have been shown to have a high predictive value of suicide rates and secondary mood and psychiatric disturbances [257]. Conventional data suggest that at least 25% of adolescents have a psychiatric disorder. This by far underestimates the problem, which is that adolescents have closer to 50% Axis 1 diagnoses and ~90% have an Axis 2 trait. The continued dumping of SUD patients to psychiatrists demonstrates the current health care system’s failure to coordinate brain health and medical comorbidities [36].

9. Neurological Imaging

SUD should be recognized as a brain disorder, and thus, neurological imaging should be implemented in its diagnosis and treatment. It is known that imaging is a very useful tool to help understand neurotransmitter interaction at regions of interest (ROI) in the brain, and over the past 40 years, electrophysiology has become the standard and most cost-effective method (Table 7) [258,259,260,261,262,263,264]. In fact, of all imaging methods, decreased p300 amplitude/latency, increased theta wave on qEEG, and abnormal evoked potential on visual and auditory processing are most established with electrophysiology (Table 7) [257,264,265,266,267,268,269,270,271,272]. Therefore, in terms of imaging, Quantitative Electroencephalogram (qEEG)/P300/Evoked Potential (EP) should become the standard in the diagnosis/treatment of SUD patients.
Long-term successful treatment in medicine is accomplished with precision biological markers [257,278,279]. When all practitioners use the same test, e.g., EKG, echocardiogram, and cholesterol, etc., we obtain the best results [280,281,282]. The most common neuropsychiatric diagnosis in the US is depression, which only worsens with SUD [257,265,266]. Our laboratory showed that patients with both depression and SUD synergistically induce an exacerbated infraction in P300 metrics (Figure 1). Since SUD is a neurological disorder, talk therapy is an adjunct to spiritual and rehab therapeutics [182,259,260,261,262,263,264,283,284,285,286,287].

10. Changing the Metric Associated with SUD: Are We Going to the Promised Land

Addiction is a substantial health issue with limited treatment options approved by the FDA and, as such, currently available. It is known that cognitive circuitry and reward circuity overlap in the brain and share similar “light switches” [292]. The advent of neuroimaging techniques that link neurochemical and neurogenetic mechanisms to the reward circuitry brain function provides a framework for potential genomic-based therapies [293].
A search of the current literature (9-26-21) via PUBMED provides descriptions of promising new targets, alternatives such as animal models of gene therapy, addiction treatment and relapse prevention, and nutrigenomic and pharmacogenomic methods to manipulate transcription and gene expression [294]. In our opinion, while developing a cost-effective methodology to help primary care physicians incorporate the proposed BHC, we recognize the clinical benefit of early genetic testing to determine addiction risk stratification and dopaminergic agonistic (in subjects with hypodopaminergia) and antagonistic (in subjects with hyperdopaminergia) therapies. Adoption of these actionable targets, especially in the promotion of precision medicine, is potentially the genome-based wave of the future. In addition, further development, especially in gene transfer work (gene editing) and viral vector identification, could be the future of gene therapy for RDS. This could be accomplished through candidate and genome-wide association studies. Many gene clusters and polymorphisms implicated in drug, food, and behavioral dependence are linked by the common rubric RDS [293,294].

11. Risk Assessment Instruments to Identify Cognitive Brain Dysfunction

Since SUD is widely correlated with neuropsychiatric, medical, and social consequences, methods are reviewed to identify comorbidities and their high risk (Table 1, Table 2, Table 4 and Table 5) [36]. One very critical area of assessment must include a detailed family history of familiar SUD. The premorbid risk factors include virtually any neuropsychiatric/sleep disorders, traumatic brain injury, concussion, head trauma, family history of addiction, neurotransmitter and second messenger polymorphisms, hormonal abnormalities, and other areas of dysfunction (Table 2). Early identification of genetic risk for all reward dysregulation, either deficit or surfeit in terms of dopaminergic activity, is critical prior to any initiation of unwanted substances or even non-substance seeking as a preventive step [295].
Dopamine genotyping is critical for pediatrics because “an increase in dopamine levels is typical of all addictive drugs (final common SUD pathway) and sufficient to trigger forms of synaptic plasticity underlying adaptive behaviors” [296,297,298]. Genotyping predicts a higher probability of SUD in pediatric patients, but SUD is not completely determined by genes at any age [196,294,295,296]. Genes are a risk factor, particularly when young, but as individuals age, they transcend genetics with epigenetics. Therefore, the BHC is more essential long term.

12. Gateway to Reward Dysregulation: Reward Gene Antecedents to Abnormal Cravings

Drug and alcohol experimentation is almost endless, from bath salts to psychedelics (Table 8) [299,300,301,302,303]. Currently, despite the existence of the GARS test and possibly other viable genetically based panels, most clinicians involved in addiction medicine fail to identify SUD appropriately and objectively in patients of all ages. This conundrum translates to lack of psychometrics for evaluating the premorbid risks of SUD as part of primary care standard medicine [180].
Subjective medical histories of SUD and even medical patients alone are unreliable; for example, patients misreport their current height by 0.5–4 inches [333,334,335,336,337,338]. Testing of reward dysregulation such as SUD is marked by underreporting of patients not willing to testify against themselves, and moreover, there is difficulty in identifying the number of substances being used through standard testing, e.g., urine drug screens, breathalyzers, blood alcohol levels, etc. It is known that, for example, patients that carry the DRD2 A1 allele are more likely to deny or lie about their SUD issue [338]. Specifically, Comings et al. investigated the dopamine D2 receptor (DRD2) gene haplotypes, identified by the allele-specific polymerase chain reaction of two mutations (G/T and C/T) 241 base pairs apart, in 57 of the ATU subjects and 42 of the controls [338]. They reported that subjects with the one haplotype tended to show a decrease in mature and an increase in neurotic and immature defense styles compared to those without the one haplotype. These results suggest that the DRD2 locus is one factor controlling defense styles (lying).
The pediatric population will also benefit from a BHC. Currently, there is inadequate subjective and virtually no objective psychometrics to analyze the premorbid risks of SUD in the pediatric population (Table 3). However, according to NCDAS, ~50% of teenagers have experimented with illicit drugs at least once [12]. Early drug use/abuse has been shown to correlate with SUD later in life, and according to the gateway hypothesis, this early experimentation can escalate into more addictive illicit drugs later in adulthood [12,300,301,339]. Each episode of experimentation likely injures the brain to some degree, and eventually, these injuries accumulate, resulting in SUD [299,300,301,340]. Of interest in terms of a gateway hypothesis to SUD, Kandel and Kandel [339] describe how marijuana and other illicit drug use are preceded by tobacco or alcohol use. Additionally, using their mouse model, they were able to identify biological processes, showing that nicotine is a gateway drug that exerts a priming effect on cocaine through epigenetically increased global acetylation in the striatum [339].
It is noteworthy that it may be difficult in some cases to misdiagnose SUD with other conditions, and even careful following of DSM-5 may not provide the best possible manner to determine SUD compared with other neurological diseases. However, with the coupling of a strong family history and genetic addiction risk assessment, this might be a way to prevent misdiagnosis. Nevertheless, if we could agree that SUD is just a subset of a more major umbrella terminology such as reward deficiency, then it may not be such a real issue due to overall common reward circuitry dysregulation in general.

13. Conclusions

Once a person is in rehab or detox, it is the equivalent of their first stroke or heart attack, and by then, treatment is too late. The tools to prevent the progression of SUD are available and must be utilized; otherwise, the deaths attributed to addiction will continue to rise. The approach to brain health and addiction needs to be reexamined, and a new approach must be implemented by the medical community as a whole. At least three decades of clinical research reviewed here establishes a “Brain Health Check” based on precision medicine for the SUD phenotype centered around precision neuropsychiatric testing, i.e., Memory (CNSVS), Attention (TOVA), Neuropsychiatric (MCMI-III), Neurological Imaging (qEEG/P300/EP). The BHC establishes a set of objective assessments for brain health that parallel those used to treat cardiac disease and provides a form of standardization that is desperately needed. This approach would greatly improve the brain/mind health of all and help stop/prevent the rise of SUD with early detection and treatment, thus bringing an attenuation to the current addiction pandemic.
Moreover, The Carter Center has estimated that if the addiction crisis continues at the same rate, by the year 2030, it will cost America approximately 16 trillion dollars. The current status of our precious youth has been compromised by not only the COVID pandemic but also by the unfortunate state of an unwanted global opioid crisis, especially in America. Hundreds of thousands of people have succumbed to deadly opioid type drugs, a rate that is increasing yearly. The neurodevelopment of our children has been compromised by not only the victimization of mothers using opioids and other drugs during pregnancy but also by a high rate of DNA polymorphic antecedents as well as methylation on specific important genes related to normal brain function via known epigenetic insults. Along with these genetic antecedent insults affecting normal mRNA transcription and loss of required proteins to induce normal brain function, normal brain development in our youth is very complex.
It is generally accepted that myelination in the frontal cortex is quite delayed in our youth, especially as it relates to executive function and decision making. However, we embrace positive thinking, especially as it relates to neurotheology. An understanding of this short circuiting in brain development, along with potential high antecedent polymorphic risk variants or alleles and generational epigenetics, provides a clear rationale to embrace the Brain Research Commission (BRC) suggestion to mimic fitness programs with an adaptable Brain Health Check-up. This can be implemented in America and other countries’ educational systems. Adoption of this proposal may reduce juvenile criminal activities and potential attenuation of relapse to RDS behaviors.

Author Contributions

Conceptualization, E.R.B.; writing—original draft preparation, E.R.B., C.A.D. and K.B.; writing—review and editing, M.S.G., A.B., A.G., D.B., A.K.R., D.E.S. and J.L.C.; supervision, K.B.; project administration, A.G. All authors have read and agreed to the published version of the manuscript.

Funding

Funding was provided by The Kenneth Blum Institute on Behavior and Neurogenetics, Austin, TX. 78701, USA, and NIDA intramural program for JLC.

Conflicts of Interest

K.B. is the inventor of GARS and KB220 variants and is credited with domestic and foreign issued and pending patents. K.B. has entered into an exclusive licensing agreement with Ivitalize, Inc. The other authors declare no conflict of interest.

References

  1. Csete, J.; Kamarulzaman, A.; Kazatchkine, M.; Altice, F.; Balicki, M.; Buxton, J.; Cepeda, J.; Comfort, M.; Goosby, E.; Goulão, J.; et al. Public Health and International Drug Policy. Lancet 2016, 387, 1427–1480. [Google Scholar] [CrossRef] [Green Version]
  2. American Psychiatric Association. Diagnostic & Statistical Manual of Mental Disorders (Dsm-IV), 4th ed.; American Psychiatric Publishing: Arlington, TX, USA, 1994. [Google Scholar]
  3. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders (DSM-5 (R)), 5th ed.; American Psychiatric Association Publishing: Arlington, TX, USA, 2013. [Google Scholar]
  4. Substance Abuse and Mental Health Services Administration. Mental Health and Substance Use Disorders. Available online: https://www.samhsa.gov/find-help/disorders (accessed on 28 September 2021).
  5. Substance Abuse and Mental Health Services Administration. Opioid Overdose. Available online: https://www.samhsa.gov/medication-assisted-treatment/medications-counseling-related-conditions/opioid-overdose (accessed on 28 September 2021).
  6. National Institute on Drug Abuse. The Science of Drug Use and Addiction: The basics. Available online: https://www.drugabuse.gov/publications/media-guide/science-drug-use-addiction-basics (accessed on 28 September 2021).
  7. National Institute on Alcohol Abuse and Alcoholism. Understanding Alcohol Use Disorder. Available online: https://www.niaaa.nih.gov/publications/brochures-and-fact-sheets/understanding-alcohol-use-disorder (accessed on 28 September 2021).
  8. National Institute on Drug Abuse. Is Marijuana a gateway drug? Available online: https://www.drugabuse.gov/publications/research-reports/marijuana/marijuana-gateway-drug (accessed on 28 September 2021).
  9. Bustamante, J. NCDAS: Substance Abuse and Addiction Statistics 2021. Available online: https://drugabusestatistics.org/ (accessed on 27 September 2021).
  10. National Center for Drug Abuse Statistics. Drug Use Among Youth: Facts & Statistics. Available online: https://drugabusestatistics.org/teen-drug-use/ (accessed on 2 February 2022).
  11. National Institute on Drug Abuse. Monitoring the Future Study: Trends in Prevalence of Various Drugs. Available online: https://www.drugabuse.gov/drug-topics/trends-statistics/monitoring-future/monitoring-future-study-trends-in-prevalence-various-drugs (accessed on 1 October 2021).
  12. Substance Abuse and Mental Health Services Administration. NSDUH Annual National Report. 2020. Available online: https://www.samhsa.gov/data/report/2020-nsduh-annual-national-report (accessed on 23 April 2022).
  13. Sudhinaraset, M.; Wigglesworth, C.; Takeuchi, D.T. Social and Cultural Contexts of Alcohol Use: Influences in a Social-Ecological Framework. Alcohol Res. 2016, 38, 35–45. [Google Scholar] [PubMed]
  14. National Institute on Drug Abuse. Alcohol. Available online: https://nida.nih.gov/drug-topics/alcohol#Ref (accessed on 23 April 2022).
  15. Grant, B.F.; Goldstein, R.B.; Saha, T.D.; Chou, S.P.; Jung, J.; Zhang, H.; Pickering, R.P.; Ruan, W.J.; Smith, S.M.; Huang, B.; et al. Epidemiology of DSM-5 Alcohol Use Disorder: Results from the National Epidemiologic Survey on Alcohol and Related Conditions III. JAMA Psychiatry 2015, 72, 757–766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Cocaine Use in Europe 2019. Available online: https://www.statista.com/statistics/597731/cocaine-use-europe-by-country/ (accessed on 2 October 2021).
  17. Topic: Drug Situation in Europe. Available online: https://www.statista.com/topics/3823/drug-situation-in-europe/ (accessed on 2 October 2021).
  18. Addiction in the EU. Available online: https://www.addictioncenter.com/addiction/addiction-in-the-eu/ (accessed on 2 October 2021).
  19. Degenhardt, L.; Bharat, C.; Glantz, M.D.; Sampson, N.A.; Al-Hamzawi, A.; Alonso, J.; Andrade, L.H.; Bunting, B.; Cia, A.; de Girolamo, G.; et al. Association of Cohort and Individual Substance Use with Risk of Transitioning to Drug Use, Drug Use Disorder, and Remission from Disorder: Findings from the World Mental Health Surveys: Findings from the World Mental Health Surveys. JAMA Psychiatry 2019, 76, 708–720. [Google Scholar] [CrossRef]
  20. Dhawan, A.; Rao, R.; Ambekar, A.; Pusp, A.; Ray, R. Treatment of Substance Use Disorders through the Government Health Facilities: Developments in the “Drug De-Addiction Programme” of Ministry of Health and Family Welfare, Government of India. Indian J. Psychiatry 2017, 59, 380–384. [Google Scholar]
  21. Pacurucu-Castillo, S.F.; Ordóñez-Mancheno, J.M.; Hernández-Cruz, A.; Alarcón, R.D. World Opioid and Substance Use Epidemic: A Latin American Perspective. Psychiatr. Res. Clin. Pract. 2019, 1, 32–38. [Google Scholar] [CrossRef] [Green Version]
  22. International Society of Substance Use Professionals. China. Available online: https://www.issup.net/knowledge-share/country-profiles/china (accessed on 2 October 2021).
  23. Risgaard, B. Sudden Cardiac Death: A Nationwide Cohort Study among the Young. Dan. Med. J. 2016, 63, B5321. [Google Scholar]
  24. Mинаева, A.; Bайсман, К. The peculiarities of coding and the determination of the primary cause of death from the diseases induced by the human immunodeficiency virus in accordance with ICD-10. Sud. Med. Ekspert. 2015, 58, 27–29. [Google Scholar] [CrossRef]
  25. Nielsen, G.P.; Björnsson, J.; Jonasson, J.G. The Accuracy of Death Certificates: Implications for Health Statistics. Vichows Archiv A Pathol Anat 1991, 419, 143–146. [Google Scholar] [CrossRef]
  26. Povzun, S.A. About eligibility of certificates of death from hepatic insufficiency. Sud. Med. Ekspert. 2013, 56, 48–49. [Google Scholar]
  27. Wogen, J.; Restrepo, M.T. Human Rights, Stigma, and Substance Use. Health Hum. Rights 2020, 22, 51–60. [Google Scholar] [PubMed]
  28. Davis, C.G.; Doherty, S.; Moser, A.E. Social Desirability and Change Following Substance Abuse Treatment in Male Offenders. Psychol. Addict. Behav. 2014, 28, 872–879. [Google Scholar] [CrossRef] [PubMed]
  29. Clark, C.B.; Zyambo, C.M.; Li, Y.; Cropsey, K.L. The Impact of Non-Concordant Self-Report of Substance Use in Clinical Trials Research. Addict. Behav. 2016, 58, 74–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. McGinty, E.E.; Goldman, H.H.; Pescosolido, B.; Barry, C.L. Portraying Mental Illness and Drug Addiction as Treatable Health Conditions: Effects of a Randomized Experiment on Stigma and Discrimination. Soc. Sci. Med. 2015, 126, 73–85. [Google Scholar] [CrossRef]
  31. Mejia-Lancheros, C.; Lachaud, J.; O’Campo, P.; Wiens, K.; Nisenbaum, R.; Wang, R.; Hwang, S.W.; Stergiopoulos, V. Trajectories and Mental Health-Related Predictors of Perceived Discrimination and Stigma among Homeless Adults with Mental Illness. PLoS ONE 2020, 15, e0229385. [Google Scholar] [CrossRef]
  32. Kumpfer, K.L. Family-Based Interventions for the Prevention of Substance Abuse and Other Impulse Control Disorders in Girls. ISRN Addict. 2014, 2014, 308789. [Google Scholar] [CrossRef]
  33. Henry, K.L.; Knight, K.E.; Thornberry, T.P. School Disengagement as a Predictor of Dropout, Delinquency, and Problem Substance Use during Adolescence and Early Adulthood. J. Youth Adolesc. 2012, 41, 156–166. [Google Scholar] [CrossRef]
  34. Kalechstein, A.D.; De la Garza, R., II; Newton, T.F.; Green, M.F.; Cook, I.A.; Leuchter, A.F. Quantitative EEG Abnormalities Are Associated with Memory Impairment in Recently Abstinent Methamphetamine-Dependent Individuals. J. Neuropsychiatry Clin. Neurosci. 2009, 21, 254–258. [Google Scholar] [CrossRef]
  35. Peppin, J.F.; Coleman, J.J.; Paladini, A.; Varrassi, G. What Your Death Certificate Says about You May Be Wrong: A Narrative Review on CDC’s Efforts to Quantify Prescription Opioid Overdose Deaths. Cureus 2021, 13, e18012. [Google Scholar] [CrossRef]
  36. Bahorik, A.L.; Satre, D.D.; Kline-Simon, A.H.; Weisner, C.M.; Campbell, C.I. Alcohol, Cannabis, and Opioid Use Disorders, and Disease Burden in an Integrated Health Care System. J. Addict. Med. 2017, 11, 3–9. [Google Scholar] [CrossRef]
  37. Centers for Disease Control and Prevention. Leading Causes of Death. Available online: https://www.cdc.gov/nchs/fastats/leading-causes-of-death.htm (accessed on 28 September 2021).
  38. American Medical Association. Issue brief: Nation’s Drug-Related Overdose and Death Epidemic Continues to Worsen. Available online: https://www.ama-assn.org/system/files/issue-brief-increases-in-opioid-related-overdose.pdf (accessed on 27 September 2021).
  39. Centers for Disease Control and Prevention. Overdose Deaths Accelerating During COVID-19. Available online: https://www.cdc.gov/media/releases/2020/p1218-overdose-deaths-covid-19.html (accessed on 27 September 2021).
  40. Centers for Disease Control and Prevention. America’s Drug Overdose Epidemic: Putting Data to Action. Available online: https://www.cdc.gov/injury/features/prescription-drug-overdose/index.html (accessed on 27 September 2021).
  41. National Institute on Drug Abuse. Overdose Death Rates. Available online: https://www.drugabuse.gov/drug-topics/trends-statistics/overdose-death-rates (accessed on 27 September 2021).
  42. National Institute on Drug Abuse. Costs of Substance Abuse. Available online: https://archives.drugabuse.gov/trends-statistics/costs-substance-abuse (accessed on 27 September 2021).
  43. Sacks, J.J.; Gonzales, K.R.; Bouchery, E.E.; Tomedi, L.E.; Brewer, R.D. 2010 National and State Costs of Excessive Alcohol Consumption. Am. J. Prev. Med. 2015, 49, e73–e79. [Google Scholar] [CrossRef] [PubMed]
  44. National Institute on Alcohol Abuse and Alcoholism. Alcohol Facts and Statistics. Available online: https://www.niaaa.nih.gov/publications/brochures-and-fact-sheets/alcohol-facts-and-statistics (accessed on 27 September 2021).
  45. World Health Organization. Alcohol and Drug Use Disorders: Global Health Estimates. Available online: https://www.who.int/substance_abuse/activities/fadab/msb_adab_2017_GHE_23June2017.pdf (accessed on 28 September 2021).
  46. Substance Abuse and Mental Health Services Administration. Substance Abuse Prevention Dollars and Cents: A Cost-Benefit Analysis. Available online: https://www.samhsa.gov/sites/default/files/cost-benefits-prevention.pdf (accessed on 28 September 2021).
  47. Sunstein, C.R.; Posner, E.A. Dollars and Death. SSRN Electron. J. 2004, 72, 537. [Google Scholar] [CrossRef] [Green Version]
  48. Blum, K.; Gardner, E.; Oscar-Berman, M.; Gold, M. “Liking” and “Wanting” Linked to Reward Deficiency Syndrome (RDS): Hypothesizing Differential Responsivity in Brain Reward Circuitry. Curr. Pharm. Des. 2012, 18, 113–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Wu, L.-T.; Ghitza, U.E.; Zhu, H.; Spratt, S.; Swartz, M.; Mannelli, P. Substance Use Disorders and Medical Comorbidities among High-Need, High-Risk Patients with Diabetes. Drug Alcohol Depend. 2018, 186, 86–93. [Google Scholar] [CrossRef] [PubMed]
  50. Winkelman, T.N.A.; Villapiano, N.; Kozhimannil, K.B.; Davis, M.M.; Patrick, S.W. Incidence and Costs of Neonatal Abstinence Syndrome among Infants with Medicaid: 2004–2014. Pediatrics 2018, 141, e20173520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Ladha, K.S.; Zhao, K.; Quraishi, S.A.; Kurth, T.; Eikermann, M.; Kaafarani, H.M.A.; Klein, E.N.; Seethala, R.; Lee, J. The Deyo-Charlson and Elixhauser-van Walraven Comorbidity Indices as Predictors of Mortality in Critically Ill Patients. BMJ Open 2015, 5, e008990. [Google Scholar] [CrossRef] [PubMed]
  52. Hall, O.T.; Trimble, C.; Garcia, S.; Entrup, P.; Deaner, M.; Teater, J. Unintentional Drug Overdose Mortality in Years of Life Lost among Adolescents and Young People in the US from 2015 to 2019. JAMA Pediatr. 2022, 176, 415–417. [Google Scholar] [CrossRef]
  53. Blum, K.; Chen, T.J.H.; Chen, A.L.H.; Madigan, M.; Downs, B.W.; Waite, R.L.; Braverman, E.R.; Kerner, M.; Bowirrat, A.; Giordano, J.; et al. Do Dopaminergic Gene Polymorphisms Affect Mesolimbic Reward Activation of Music Listening Response? Therapeutic Impact on Reward Deficiency Syndrome (RDS). Med. Hypotheses 2010, 74, 513–520. [Google Scholar] [CrossRef]
  54. Alves, M.R.P.; Yamamoto, T.; Arias-Carrión, O.; Rocha, N.B.F.; Nardi, A.E.; Machado, S.; Silva, A.C. Executive Function Impairments in Patients with Depression. CNS Neurol. Disord. Drug Targets 2014, 13, 1026–1040. [Google Scholar] [CrossRef] [Green Version]
  55. Lepage, C.; Smith, A.M.; Moreau, J.; Barlow-Krelina, E.; Wallis, N.; Collins, B.; MacKenzie, J.; Scherling, C. A Prospective Study of Grey Matter and Cognitive Function Alterations in Chemotherapy-Treated Breast Cancer Patients. Springerplus 2014, 3, 444. [Google Scholar] [CrossRef] [Green Version]
  56. Frieri, G.; Galletti, B.; Serva, D.; Viscido, A. The Role on Endoscopy in Alcohol-Related Diseases. Rev. Recent Clin. Trials 2016, 11, 196–200. [Google Scholar] [CrossRef] [PubMed]
  57. Griffin, M.R.; Piper, J.M.; Daugherty, J.R.; Snowden, M.; Ray, W.A. Nonsteroidal Anti-Inflammatory Drug Use and Increased Risk for Peptic Ulcer Disease in Elderly Persons. Ann. Intern. Med. 1991, 114, 257–263. [Google Scholar] [CrossRef]
  58. Irving, H.M.; Samokhvalov, A.V.; Rehm, J. Alcohol as a Risk Factor for Pancreatitis. A Systematic Review and Meta-Analysis. JOP 2009, 10, 387–392. [Google Scholar] [PubMed]
  59. Torres, J.-L.; Novo-Veleiro, I.; Manzanedo, L.; Alvela-Suárez, L.; Macías, R.; Laso, F.-J.; Marcos, M. Role of MicroRNAs in Alcohol-Induced Liver Disorders and Non-Alcoholic Fatty Liver Disease. World J. Gastroenterol. 2018, 24, 4104–4118. [Google Scholar] [CrossRef] [PubMed]
  60. Dubinkina, V.B.; Tyakht, A.V.; Odintsova, V.Y.; Yarygin, K.S.; Kovarsky, B.A.; Pavlenko, A.V.; Ischenko, D.S.; Popenko, A.S.; Alexeev, D.G.; Taraskina, A.Y.; et al. Links of Gut Microbiota Composition with Alcohol Dependence Syndrome and Alcoholic Liver Disease. Microbiome 2017, 5, 141. [Google Scholar] [CrossRef] [PubMed]
  61. Li, J.; Niu, J.-Z.; Wang, J.-F.; Li, Y.; Tao, X.-H. Pathological Mechanisms of Alcohol-Induced Hepatic Portal Hypertension in Early Stage Fibrosis Rat Model. World J. Gastroenterol. 2005, 11, 6483–6488. [Google Scholar] [CrossRef]
  62. Hutchinson, D.R.; Halliwell, R.P.; Lockhart, J.D.F.; Parke, D.V. Glycylprolyl-p-Nitroanilidase in Hepatobiliary Disease. Clin. Chim. Acta 1981, 109, 83–89. [Google Scholar] [CrossRef]
  63. Conn, H.O. Spontaneous Bacterial Peritonitis: Variant Syndromes. South. Med. J. 1987, 80, 1343–1346. [Google Scholar] [CrossRef]
  64. Schneider, A.; Hirth, M.; Weiss, C.; Weidner, P.; Antoni, C.; Thomann, A.; Reindl, W.; Ebert, M.P.; Pfützer, R.H. Prevalence of Inflammatory Bowel Disease in Alcoholic, Non-Alcoholic and Autoimmune Pancreatitis. Z. Gastroenterol. 2018, 56, 469–478. [Google Scholar]
  65. Barritt, A.S., IV; Lee, B.; Runge, T.; Schmidt, M.; Jhaveri, R. Increasing Prevalence of Hepatitis C among Hospitalized Children Is Associated with an Increase in Substance Abuse. J. Pediatr. 2018, 192, 159–164. [Google Scholar] [CrossRef] [Green Version]
  66. Ruotsalainen, E.; Kardén-Lilja, M.; Kuusela, P.; Vuopio-Varkila, J.; Virolainen-Julkunen, A.; Sarna, S.; Valtonen, V.; Järvinen, A. Methicillin-Sensitive Staphylococcus Aureus Bacteraemia and Endocarditis among Injection Drug Users and Nonaddicts: Host Factors, Microbiological and Serological Characteristics. J. Infect. 2008, 56, 249–256. [Google Scholar] [CrossRef] [PubMed]
  67. Sahuquillo-Arce, J.M.; Menéndez, R.; Méndez, R.; Amara-Elori, I.; Zalacain, R.; Capelastegui, A.; Aspa, J.; Borderías, L.; Martín-Villasclaras, J.J.; Bello, S.; et al. Age-Related Risk Factors for Bacterial Aetiology in Community-Acquired Pneumonia: Pathogens, Comorbidities and Pneumonia. Respirology 2016, 21, 1472–1479. [Google Scholar] [CrossRef] [PubMed]
  68. Ricks, P.M.; Hershow, R.C.; Rahimian, A.; Huo, D.; Johnson, W.; Prachand, N.; Jimenez, A.; Wiebel, W.; Paul, W. A Randomized Trial Comparing Standard Outcomes in Two Treatment Models for Substance Users with Tuberculosis. Int. J. Tuberc. Lung Dis. 2015, 19, 326–332. [Google Scholar] [CrossRef] [PubMed]
  69. Huang, D.B.; Noviello, S.; Balser, B.; Scaramucci, A.; Corey, G.R. A Pooled Analysis of the Safety and Efficacy of Iclaprim versus Vancomycin for the Treatment of Acute Bacterial Skin and Skin Structure Infections in Patients with Intravenous Drug Use: Phase 3 REVIVE Studies. Clin. Ther. 2019, 41, 1090–1096. [Google Scholar] [CrossRef]
  70. Cordova, D.; Munoz-Velazquez, J.; Mendoza Lua, F.; Fessler, K.; Warner, S.; Delva, J.; Adelman, N.; Fernandez, A.; Bauermeister, J.; Youth Leadership Council. Pilot Study of a Multilevel Mobile Health App for Substance Use, Sexual Risk Behaviors, and Testing for Sexually Transmitted Infections and HIV among Youth: Randomized Controlled Trial. JMIR MHealth UHealth 2020, 8, e1625. [Google Scholar] [CrossRef]
  71. Dale, S.K.; Traeger, L.; O’Cleirigh, C.; Bedoya, C.A.; Pinkston, M.; Wilner, J.G.; Stein, M.; Safren, S.A. Baseline Substance Use Interferes with Maintenance of HIV Medication Adherence Skills. AIDS Patient Care STDS 2016, 30, 215–220. [Google Scholar] [CrossRef]
  72. Khanna, D.; Clements, P.J.; Furst, D.E.; Korn, J.H.; Ellman, M.; Rothfield, N.; Wigley, F.M.; Moreland, L.W.; Silver, R.; Kim, Y.H.; et al. Recombinant Human Relaxin in the Treatment of Systemic Sclerosis with Diffuse Cutaneous Involvement: A Randomized, Double-Blind, Placebo-Controlled Trial. Arthritis Rheum. 2009, 60, 1102–1111. [Google Scholar] [CrossRef] [Green Version]
  73. Zhu, J.; Nelson, K.; Toth, J.; Muscat, J.E. Nicotine Dependence as an Independent Risk Factor for Atherosclerosis in the National Lung Screening Trial. BMC Public Health 2019, 19, 103. [Google Scholar] [CrossRef] [Green Version]
  74. Jaladi, P.R.; Patel, V.; Kuduva Rajan, S.; Rashid, W.; Madireddy, S.; Ajibawo, T.; Imran, S.; Patel, R.S. Arrhythmia-Related Hospitalization and Comorbid Cannabis Use Disorder: Trend Analysis in US Hospitals (2010–2014). Cureus 2019, 11, e5607. [Google Scholar] [CrossRef] [Green Version]
  75. Kueh, S.-H.A.; Gabriel, R.S.; Lund, M.; Sutton, T.; Bradley, J.; Kerr, A.J.; Looi, J.-L. Clinical Characteristics and Outcomes of Patients with Amphetamine-Associated Cardiomyopathy in South Auckland, New Zealand. Heart Lung Circ. 2016, 25, 1087–1093. [Google Scholar] [CrossRef]
  76. Pinn, G.; Bovet, P. Alcohol-related Cardiomyopathy in the Seychelles. Med. J. Aust. 1991, 155, 529–532. [Google Scholar] [CrossRef] [PubMed]
  77. Kelbaek, H.; Heslet, L.; Skagen, K.; Munck, O.; Christensen, N.J.; Godtfredsen, J. Cardiac Function after Alcohol Ingestion in Patients with Ischemic Heart Disease and Cardiomyopathy: A Controlled Study. Alcohol Alcohol. 1988, 23, 17–21. [Google Scholar] [PubMed]
  78. Mau, M.K.; Asao, K.; Efird, J.; Saito, E.; Ratner, R.; Hafi, M.; Seto, T. Risk Factors Associated with Methamphetamine Use and Heart Failure among Native Hawaiians and Other Pacific Island Peoples. Vasc. Health Risk Manag. 2008, 5, 45. [Google Scholar] [CrossRef] [Green Version]
  79. Lin, S.; Leppla, I.E.; Yan, H.; Probert, J.M.; Randhawa, P.A.; Leoutsakos, J.-M.S.; Probasco, J.C.; Neufeld, K.J. Prevalence and Improvement of Caine-Positive Wernicke-Korsakoff Syndrome in Psychiatric Inpatient Admissions Psychosomatics. Psychosomatics 2020, 2020, 31–38. [Google Scholar] [CrossRef] [PubMed]
  80. Bowe, A.; Rosenheck, R. PTSD and Substance Use Disorder among Veterans: Characteristics, Service Utilization and Pharmacotherapy. J. Dual Diagn. 2015, 11, 22–32. [Google Scholar] [CrossRef] [PubMed]
  81. Winhusen, T.; Theobald, J.; Kaelber, D.C.; Lewis, D. Medical Complications Associated with Substance Use Disorders in Patients with Type 2 Diabetes and Hypertension: Electronic Health Record Findings. Addiction 2019, 114, 1462–1470. [Google Scholar] [CrossRef]
  82. Robinson-Papp, J.; Gelman, B.B.; Grant, I.; Singer, E.; Gensler, G.; Morgello, S.; for the National NeuroAIDS Tissue Consortium. Substance Abuse Increases the Risk of Neuropathy in an HIV-Infected Cohort: Substance Use and HIV Neuropathy. Muscle Nerve 2012, 45, 471–476. [Google Scholar] [CrossRef] [Green Version]
  83. Tapper, E.B.; Parikh, N.D.; Green, P.K.; Berry, K.; Waljee, A.K.; Moon, A.M.; Ioannou, G.N. Reduced Incidence of Hepatic Encephalopathy and Higher Odds of Resolution Associated with Eradication of HCV Infection. Clin. Gastroenterol. Hepatol. 2020, 18, 1197–1206.e7. [Google Scholar] [CrossRef]
  84. Bernick, C.; Banks, S.J.; Shin, W.; Obuchowski, N.; Butler, S.; Noback, M.; Phillips, M.; Lowe, M.; Jones, S.; Modic, M. Repeated Head Trauma Is Associated with Smaller Thalamic Volumes and Slower Processing Speed: The Professional Fighters’ Brain Health Study. Br. J. Sports Med. 2015, 49, 1007–1011. [Google Scholar] [CrossRef]
  85. Mihalik, J.P.; Lengas, E.; Register-Mihalik, J.K.; Oyama, S.; Begalle, R.L.; Guskiewicz, K.M. The Effects of Sleep Quality and Sleep Quantity on Concussion Baseline Assessment. Clin. J. Sport Med. 2013, 23, 343–348. [Google Scholar] [CrossRef]
  86. Bernstein, G.A.; Carroll, M.E.; Crosby, R.D.; Perwien, A.R.; Go, F.S.; Benowitz, N.L. Caffeine Effects on Learning, Performance, and Anxiety in Normal School-Age Children. J. Am. Acad. Child Adolesc. Psychiatry 1994, 33, 407–415. [Google Scholar] [CrossRef] [PubMed]
  87. Goez, H.; Back-Bennet, O.; Zelnik, N. Differential Stimulant Response on Attention in Children with Comorbid Anxiety and Oppositional Defiant Disorder. J. Child Neurol. 2007, 22, 538–542. [Google Scholar] [CrossRef] [PubMed]
  88. Onnink, A.M.H.; Zwiers, M.P.; Hoogman, M.; Mostert, J.C.; Kan, C.C.; Buitelaar, J.; Franke, B. Brain Alterations in Adult ADHD: Effects of Gender, Treatment and Comorbid Depression. Eur. Neuropsychopharmacol. 2014, 24, 397–409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Herr, J.; Hatch, L.; Sephien, A.; Hanna, K. 27-Year-Old Woman • Postpartum Seizures • PTSD • History of Depression • Dx? J. Fam. Pract. 2021, 70, 300–302. [Google Scholar] [CrossRef]
  90. Xu, L.; Nan, J.; Lan, Y. The Nucleus Accumbens: A Common Target in the Comorbidity of Depression and Addiction. Front. Neural Circuits 2020, 14, 37. [Google Scholar] [CrossRef]
  91. Maremmani, A.G.I.; Bacciardi, S.; Gehring, N.D.; Cambioli, L.; Schütz, C.; Jang, K.; Krausz, M. Substance Use among Homeless Individuals with Schizophrenia and Bipolar Disorder. J. Nerv. Ment. Dis. 2017, 205, 173–177. [Google Scholar] [CrossRef]
  92. Gold, A.K.; Peters, A.T.; Otto, M.W.; Sylvia, L.G.; da Silva Magalhaes, P.V.; Berk, M.; Dougherty, D.D.; Miklowitz, D.J.; Frank, E.; Nierenberg, A.A.; et al. The Impact of Substance Use Disorders on Recovery from Bipolar Depression: Results from the Systematic Treatment Enhancement Program for Bipolar Disorder Psychosocial Treatment Trial. Aust. N. Z. J. Psychiatry 2018, 52, 847–855. [Google Scholar] [CrossRef]
  93. Gualtieri, C.T.; Johnson, L.G. ADHD: Is Objective Diagnosis Possible? Psychiatry 2005, 2, 44–53. [Google Scholar]
  94. Ünsel-Bolat, G.; Ercan, E.S.; Bolat, H.; Süren, S.; Bacanlı, A.; Yazıcı, K.U.; Rohde, L.A. Comparisons between Sluggish Cognitive Tempo and ADHD-Restrictive Inattentive Presentation Phenotypes in a Clinical ADHD Sample. Atten. Defic. Hyperact. Disord. 2019, 11, 363–372. [Google Scholar] [CrossRef]
  95. Cook, N.E.; Huang, D.S.; Silverberg, N.D.; Brooks, B.L.; Maxwell, B.; Zafonte, R.; Berkner, P.D.; Iverson, G.L. Baseline Cognitive Test Performance and Concussion-like Symptoms among Adolescent Athletes with ADHD: Examining Differences Based on Medication Use. Clin. Neuropsychol. 2017, 31, 1341–1352. [Google Scholar] [CrossRef]
  96. Brandley, E.T.; Holton, K.F. Breakfast Positively Impacts Cognitive Function in College Students with and without ADHD. Am. J. Health Promot. 2020, 34, 668–671. [Google Scholar] [CrossRef] [PubMed]
  97. Rotem, A.; Danieli, Y.; Ben-Sheetrit, J.; Bashari, A.; Golubchik, P.; Ben-Hayun, R.; Weizman, A.; Manor, I. Apparent Lack of Practice Effects in the Test of Variables of Attention (TOVA) in Adult ADHD. Atten. Defic. Hyperact. Disord. 2019, 11, 73–81. [Google Scholar] [CrossRef] [PubMed]
  98. Jensen, P.S.; Kenny, D.T. The Effects of Yoga on the Attention and Behavior of Boys with Attention-Deficit/ Hyperactivity Disorder (ADHD). J. Atten. Disord. 2004, 7, 205–216. [Google Scholar] [CrossRef] [PubMed]
  99. Katz, M.; Levine, A.A.; Kol-Degani, H.; Kav-Venaki, L. A Compound Herbal Preparation (CHP) in the Treatment of Children with ADHD: A Randomized Controlled Trial. J. Atten. Disord. 2010, 14, 281–291. [Google Scholar] [CrossRef] [PubMed]
  100. Kollins, S.H.; DeLoss, D.J.; Cañadas, E.; Lutz, J.; Findling, R.L.; Keefe, R.S.E.; Epstein, J.N.; Cutler, A.J.; Faraone, S.V. A Novel Digital Intervention for Actively Reducing Severity of Paediatric ADHD (STARS-ADHD): A Randomised Controlled Trial. Lancet Digit Health 2020, 2, e168–e178. [Google Scholar] [CrossRef]
  101. Lin, H.-Y.; Hsieh, H.-C.; Lee, P.; Hong, F.-Y.; Chang, W.-D.; Liu, K.-C. Auditory and Visual Attention Performance in Children with ADHD: The Attentional Deficiency of ADHD Is Modality Specific. J. Atten. Disord. 2017, 21, 856–864. [Google Scholar] [CrossRef] [PubMed]
  102. Lubar, J.F.; Swartwood, M.O.; Swartwood, J.N.; O’Donnell, P.H. Evaluation of the Effectiveness of EEG Neurofeedback Training for ADHD in a Clinical Setting as Measured by Changes in T.O.V.A. Scores, Behavioral Ratings, and WISC-R Performance. Biofeedback Self. Regul. 1995, 20, 83–99. [Google Scholar] [CrossRef]
  103. Manor, I.; Rubin, J.; Daniely, Y.; Adler, L.A. Attention Benefits after a Single Dose of Metadoxine Extended Release in Adults with Predominantly Inattentive ADHD. Postgrad. Med. 2014, 126, 7–16. [Google Scholar] [CrossRef]
  104. Gualtieri, C.T.; Johnson, L.G. Medications Do Not Necessarily Normalize Cognition in ADHD Patients. J. Atten. Disord. 2008, 11, 459–469. [Google Scholar] [CrossRef]
  105. Paz, Y.; Friedwald, K.; Levkovitz, Y.; Zangen, A.; Alyagon, U.; Nitzan, U.; Segev, A.; Maoz, H.; Koubi, M.; Bloch, Y. Randomised Sham-Controlled Study of High-Frequency Bilateral Deep Transcranial Magnetic Stimulation (DTMS) to Treat Adult Attention Hyperactive Disorder (ADHD): Negative Results. World J. Biol. Psychiatry 2018, 19, 561–566. [Google Scholar] [CrossRef]
  106. Rossiter, T. The Effectiveness of Neurofeedback and Stimulant Drugs in Treating AD/HD: Part II. Replication. Appl. Psychophysiol. Biofeedback 2004, 29, 233–243. [Google Scholar] [CrossRef] [PubMed]
  107. Rotem, A.; Ben-Sheetrit, J.; Newcorn, J.; Danieli, Y.; Peskin, M.; Golubchik, P.; Ben-Hayun, R.; Weizman, A.; Manor, I. The Placebo Response in Adult ADHD as Objectively Assessed by the TOVA Continuous Performance Test. J. Atten. Disord. 2021, 25, 1311–1320. [Google Scholar] [CrossRef] [PubMed]
  108. Johns Hopkins Medicine. Attention-Deficit/Hyperactivity Disorder (ADHD) in Children. Available online: https://www.hopkinsmedicine.org/health/conditions-and-diseases/adhdadd (accessed on 13 October 2021).
  109. Vardi, K. ADHD in the Elderly: An Unexpected Diagnosis. Caring Ages 2015, 16, 10. [Google Scholar] [CrossRef]
  110. Haavik, J.; Halmøy, A.; Lundervold, A.J.; Fasmer, O.B. Clinical Assessment and Diagnosis of Adults with Attention-Deficit/Hyperactivity Disorder. Expert Rev. Neurother. 2010, 10, 1569–1580. [Google Scholar] [CrossRef]
  111. Comings, D.E.; Chen, T.J.H.; Blum, K.; Mengucci, J.F.; Blum, S.H.; Meshkin, B. Neurogenetic Interactions and Aberrant Behavioral Co-Morbidity of Attention Deficit Hyperactivity Disorder (ADHD): Dispelling Myths. Theor. Biol. Med. Model. 2005, 2, 50. [Google Scholar] [CrossRef] [Green Version]
  112. Fedotovskih, A.V.; Retyunskiy, K.Y.; Petrenko, T.S.; Kublanov, V.S. The possibilities of neurostimulation (sympathetic correction) in the treatment of amnestic (Korsakoff’s) psychosis. Zh. Nevrol. Psikhiatr. Im. S S Korsakova 2018, 118, 52–59. [Google Scholar] [CrossRef]
  113. Thylstrup, B.; Schrøder, S.; Hesse, M. Psycho-Education for Substance Use and Antisocial Personality Disorder: A Randomized Trial. BMC Psychiatry 2015, 15, 283. [Google Scholar] [CrossRef] [Green Version]
  114. Dowell, D.; Haegerich, T.M.; Chou, R. CDC Guideline for Prescribing Opioids for Chronic Pain—United States, 2016. MMWR Recomm. Rep. 2016, 65, 1624–1645. [Google Scholar] [CrossRef]
  115. Poorolajal, J.; Haghtalab, T.; Farhadi, M.; Darvishi, N. Substance Use Disorder and Risk of Suicidal Ideation, Suicide Attempt and Suicide Death: A Meta-Analysis. J. Public Health (Oxf.) 2016, 38, e282–e291. [Google Scholar] [CrossRef]
  116. Hser, Y.-I.; Mooney, L.J.; Huang, D.; Zhu, Y.; Tomko, R.L.; McClure, E.; Chou, C.-P.; Gray, K.M. Reductions in Cannabis Use Are Associated with Improvements in Anxiety, Depression, and Sleep Quality, but Not Quality of Life. J. Subst. Abuse Treat. 2017, 81, 53–58. [Google Scholar] [CrossRef]
  117. Mansfield, L.; Mendoza, C.; Flores, J.; Meeves, S.G. Effects of Fexofenadine, Diphenhydramine, and Placebo on Performance of the Test of Variables of Attention (TOVA). Ann. Allergy Asthma Immunol. 2003, 90, 554–559. [Google Scholar] [CrossRef]
  118. Small, P.; Kim, H. Allergic Rhinitis. Allergy, Asthma, and Clinical Immunology. Off. J. Can. Soc. Allergy Clin. Immunol. 2011, 1, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Webb, D.R. Beclomethasone in Steroid-Dependent Asthma. Effective Therapy and Recovery of Hypothalamo-Pituitary-Adrenal Function. JAMA 1977, 238, 1508–1511. [Google Scholar] [CrossRef] [PubMed]
  120. Chandler, G.E.; Roberts, S.J.; Chiodo, L. Resilience Intervention for Young Adults with Adverse Childhood Experiences. J. Am. Psychiatr. Nurses Assoc. 2015, 21, 406–416. [Google Scholar] [CrossRef] [PubMed]
  121. Fragou, D.; Chao, M.-R.; Hu, C.-W.; Nikolaou, K.; Kovatsi, L. Global DNA Methylation Levels in White Blood Cells of Patients with Chronic Heroin Use Disorder. A Prospective Study. Toxicol. Rep. 2021, 8, 337–342. [Google Scholar] [CrossRef] [PubMed]
  122. Orum, M.H.; Kara, M.Z. Platelet to Lymphocyte Ratio (PLR) in Alcohol Use Disorder. J. Immunoassay Immunochem. 2020, 41, 184–194. [Google Scholar] [CrossRef]
  123. Walter, K.N.; Petry, N.M. Patients with Diabetes Respond Well to Contingency Management Treatment Targeting Alcohol and Substance Use. Psychol. Health Med. 2015, 20, 916–926. [Google Scholar] [CrossRef] [Green Version]
  124. Jackson, C.T.; Covell, N.H.; Drake, R.E.; Essock, S.M. Relationship between Diabetes and Mortality among Persons with Co-Occurring Psychotic and Substance Use Disorders. Psychiatr. Serv. 2007, 58, 270–272. [Google Scholar] [CrossRef]
  125. Berthoud, H.-R.; Münzberg, H.; Morrison, C.D. Blaming the Brain for Obesity: Integration of Hedonic and Homeostatic Mechanisms. Gastroenterology 2017, 152, 1728–1738. [Google Scholar] [CrossRef] [Green Version]
  126. Kushner, R. Obesity 2021: Current Clinical Management of a Chronic, Serious Disease. J. Fam. Pract. 2021, 70, S35–S40. [Google Scholar] [CrossRef]
  127. Ng, K.P.; Pascoal, T.A.; Mathotaarachchi, S.; Chan, Y.H.; Jiang, L.; Therriault, J.; Benedet, A.L.; Shin, M.; Kandiah, N.; Greenwood, C.M.T.; et al. Neuropsychiatric Symptoms Are Early Indicators of an Upcoming Metabolic Decline in Alzheimer’s Disease. Transl. Neurodegener. 2021, 10, 1. [Google Scholar] [CrossRef] [PubMed]
  128. Muller, M.; Grobbee, D.E.; den Tonkelaar, I.; Lamberts, S.W.J.; van der Schouw, Y.T. Endogenous Sex Hormones and Metabolic Syndrome in Aging Men. J. Clin. Endocrinol. Metab. 2005, 90, 2618–2623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Umhau, J.C.; Petrulis, S.G.; Diaz, R.; Biddison, J.R.; George, A.D. Hypothalamic Function in Response to 2-Deoxy-D-Glucose in Long-Term Abstinent Alcoholics. Alcohol. Clin. Exp. Res. 2001, 25, 781–786. [Google Scholar] [CrossRef]
  130. Jayaprakash, R.R. Prevalence of Osteoporosis and Proxy Clinical Indicators of Osteoporosis in Patients on Long Term Risperidone; Christian Medical College: Vellore, India, 2009. [Google Scholar]
  131. Zacny, J.P.; Paice, J.A.; Coalson, D.W. Subjective, Psychomotor, and Physiological Effects of Pregabalin Alone and in Combination with Oxycodone in Healthy Volunteers. Pharmacol. Biochem. Behav. 2012, 100, 560–565. [Google Scholar] [CrossRef] [Green Version]
  132. Tchivileva, I.E.; Hadgraft, H.; Lim, P.F.; Di Giosia, M.; Ribeiro-Dasilva, M.; Campbell, J.H.; Willis, J.; James, R.; Herman-Giddens, M.; Fillingim, R.B.; et al. Efficacy and Safety of Propranolol for Treatment of Temporomandibular Disorder Pain: A Randomized, Placebo-Controlled Clinical Trial. Pain 2020, 161, 1755–1767. [Google Scholar] [CrossRef] [PubMed]
  133. Korthuis, P.T.; King, C.; Cook, R.R.; Khuyen, T.T.; Kunkel, L.E.; Bart, G.; Nguyen, T.; Thuy, D.T.; Bielavitz, S.; Nguyen, D.B.; et al. HIV Clinic-Based Buprenorphine plus Naloxone versus Referral for Methadone Maintenance Therapy for Treatment of Opioid Use Disorder in HIV Clinics in Vietnam (BRAVO): An Open-Label, Randomised, Non-Inferiority Trial. Lancet HIV 2021, 8, e67–e76. [Google Scholar] [CrossRef]
  134. Pan, C.-S.; Ju, T.R.; Lee, C.C.; Chen, Y.-P.; Hsu, C.-Y.; Hung, D.-Z.; Chen, W.-K.; Wang, I.-K. Alcohol Use Disorder Tied to Development of Chronic Kidney Disease: A Nationwide Database Analysis. PLoS ONE 2018, 13, e0203410. [Google Scholar] [CrossRef] [Green Version]
  135. Yang, G.S.; Mi, X.; Jackson-Cook, C.K.; Starkweather, A.R.; Lynch Kelly, D.; Archer, K.J.; Zou, F.; Lyon, D.E. Differential DNA Methylation Following Chemotherapy for Breast Cancer Is Associated with Lack of Memory Improvement at One Year. Epigenetics 2020, 15, 499–510. [Google Scholar] [CrossRef]
  136. Roth, M.Y.; Elmore, J.G.; Yi-Frazier, J.P.; Reisch, L.M.; Oster, N.V.; Miglioretti, D.L. Self-Detection Remains a Key Method of Breast Cancer Detection for U.S. Women. J. Womens. Health (Larchmt.) 2011, 20, 1135–1139. [Google Scholar] [CrossRef] [Green Version]
  137. Dretsch, M.N.; Rangaprakash, D.; Katz, J.S.; Daniel, T.A.; Goodman, A.M.; Denney, T.S.; Deshpande, G. Strength and Temporal Variance of the Default Mode Network to Investigate Chronic Mild Traumatic Brain Injury in Service Members with Psychological Trauma. J. Exp. Neurosci. 2019, 13, 1179069519833966. [Google Scholar] [CrossRef] [Green Version]
  138. Jones, K.M.; Barker-Collo, S.; Parmar, P.; Starkey, N.; Theadom, A.; Ameratunga, S.; Feigin, V.L.; BIONIC study group. Trajectories in Health Recovery in the 12 Months Following a Mild Traumatic Brain Injury in Children: Findings from the BIONIC Study. J. Prim. Health Care 2018, 10, 81–89. [Google Scholar] [CrossRef] [PubMed]
  139. Plourde, V.; Brooks, B.L. Is Computerized Cognitive Testing Useful in Children and Adolescents with Moderate-to-Severe Traumatic Brain Injury? J. Int. Neuropsychol. Soc. 2017, 23, 304–313. [Google Scholar] [CrossRef] [PubMed]
  140. Dretsch, M.N.; Lange, R.T.; Katz, J.S.; Goodman, A.; Daniel, T.A.; Deshpande, G.; Denney, T.S.; Iverson, G.L.; Robinson, J.L. Examining Microstructural White Matter in Active Duty Soldiers with a History of Mild Traumatic Brain Injury and Traumatic Stress. Open Neuroimag. J. 2017, 11, 46–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Rangaprakash, D.; Deshpande, G.; Daniel, T.A.; Goodman, A.M.; Robinson, J.L.; Salibi, N.; Katz, J.S.; Denney, T.S., Jr.; Dretsch, M.N. Compromised Hippocampus-Striatum Pathway as a Potential Imaging Biomarker of Mild-Traumatic Brain Injury and Posttraumatic Stress Disorder. Hum. Brain Mapp. 2017, 38, 2843–2864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Brooks, B.L.; Low, T.A.; Daya, H.; Khan, S.; Mikrogianakis, A.; Barlow, K.M. Test or Rest? Computerized Cognitive Testing in the Emergency Department after Pediatric Mild Traumatic Brain Injury Does Not Delay Symptom Recovery. J. Neurotrauma 2016, 33, 2091–2096. [Google Scholar] [CrossRef] [PubMed]
  143. Brooks, B.L.; Daya, H.; Khan, S.; Carlson, H.L.; Mikrogianakis, A.; Barlow, K.M. Cognition in the Emergency Department as a Predictor of Recovery after Pediatric Mild Traumatic Brain Injury. J. Int. Neuropsychol. Soc. 2016, 22, 379–387. [Google Scholar] [CrossRef] [PubMed]
  144. Barker-Collo, S.; Jones, K.; Theadom, A.; Starkey, N.; Dowell, A.; McPherson, K.; Ameratunga, S.; Dudley, M.; Te Ao, B.; Feigin, V. BIONIC Research Group. Neuropsychological Outcome and Its Correlates in the First Year after Adult Mild Traumatic Brain Injury: A Population-Based New Zealand Study. Brain Inj. 2015, 29, 1604–1616. [Google Scholar] [CrossRef]
  145. Theadom, A.; Parmar, P.; Jones, K.; Barker-Collo, S.; Starkey, N.J.; McPherson, K.M.; Ameratunga, S.; Feigin, V.L.; BIONIC Research Group. Frequency and Impact of Recurrent Traumatic Brain Injury in a Population-Based Sample. J. Neurotrauma 2015, 32, 674–681. [Google Scholar] [CrossRef]
  146. Brooks, B.L.; Khan, S.; Daya, H.; Mikrogianakis, A.; Barlow, K.M. Neurocognition in the Emergency Department after a Mild Traumatic Brain Injury in Youth. J. Neurotrauma 2014, 31, 1744–1749. [Google Scholar] [CrossRef]
  147. Krishna, R.; Grinn, M.; Giordano, N.; Thirunavukkarasu, M.; Tadi, P.; Das, S. Diagnostic Confirmation of Mild Traumatic Brain Injury by Diffusion Tensor Imaging: A Case Report. J. Med. Case Rep. 2012, 6, 66. [Google Scholar] [CrossRef] [Green Version]
  148. Harch, P.G.; Andrews, S.R.; Fogarty, E.F.; Amen, D.; Pezzullo, J.C.; Lucarini, J.; Aubrey, C.; Taylor, D.V.; Staab, P.K.; Van Meter, K.W. A Phase I Study of Low-Pressure Hyperbaric Oxygen Therapy for Blast-Induced Post-Concussion Syndrome and Post-Traumatic Stress Disorder. J. Neurotrauma 2012, 29, 168–185. [Google Scholar] [CrossRef] [PubMed]
  149. Arrieux, J.P.; Cole, W.R.; Ahrens, A.P. A Review of the Validity of Computerized Neurocognitive Assessment Tools in Mild Traumatic Brain Injury Assessment. Concussion 2017, 2, CNC31. [Google Scholar] [CrossRef] [PubMed]
  150. Dezman, Z.D.W.; Gorelick, D.A.; Buchanan, L.; Soderstrom, C.A. 20-Year Mortality after Discharge in a Cohort of 1,099 Former Trauma Inpatients with and without Substance Use Disorders. Injury 2020, 51, 2930–2937. [Google Scholar] [CrossRef] [PubMed]
  151. Ilgen, M.A.; Conner, K.R.; Valenstein, M.; Austin, K.; Blow, F.C. Violent and Nonviolent Suicide in Veterans with Substance-Use Disorders. J. Stud. Alcohol Drugs 2010, 71, 473–479. [Google Scholar] [CrossRef] [PubMed]
  152. Walton, M.A.; Resko, S.; Whiteside, L.; Chermack, S.T.; Zimmerman, M.; Cunningham, R.M. Sexual Risk Behaviors among Teens at an Urban Emergency Department: Relationship with Violent Behaviors and Substance Use. J. Adolesc. Health 2011, 48, 303–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Coulton, S.; Stockdale, K.; Marchand, C.; Hendrie, N.; Billings, J.; Boniface, S.; Butler, S.; Deluca, P.; Drummond, C.; Newbury-Birch, D.; et al. Pragmatic Randomised Controlled Trial to Evaluate the Effectiveness and Cost Effectiveness of a Multi-Component Intervention to Reduce Substance Use and Risk-Taking Behaviour in Adolescents Involved in the Criminal Justice System: A Trial Protocol (RISKIT-CJS). BMC Public Health 2017, 17, 246. [Google Scholar] [CrossRef] [Green Version]
  154. Petras, H.; Kellam, S.G.; Brown, C.H.; Muthén, B.O.; Ialongo, N.S.; Poduska, J.M. Developmental Epidemiological Courses Leading to Antisocial Personality Disorder and Violent and Criminal Behavior: Effects by Young Adulthood of a Universal Preventive Intervention in First- and Second-Grade Classrooms. Drug Alcohol Depend. 2008, 95, S45–S59. [Google Scholar] [CrossRef] [Green Version]
  155. Spohr, S.A.; Livingston, M.D.; Taxman, F.S.; Walters, S.T. What’s the Influence of Social Interactions on Substance Use and Treatment Initiation? A Prospective Analysis among Substance-Using Probationers. Addict. Behav. 2019, 89, 143–150. [Google Scholar] [CrossRef]
  156. Cropsey, K.L.; Lane, P.S.; Hale, G.J.; Jackson, D.O.; Clark, C.B.; Ingersoll, K.S.; Islam, M.A.; Stitzer, M.L. Results of a Pilot Randomized Controlled Trial of Buprenorphine for Opioid Dependent Women in the Criminal Justice System. Drug Alcohol Depend. 2011, 119, 172–178. [Google Scholar] [CrossRef] [Green Version]
  157. Somers, J.M.; Moniruzzaman, A.; Palepu, A. Changes in Daily Substance Use among People Experiencing Homelessness and Mental Illness: 24-Month Outcomes Following Randomization to Housing First or Usual Care: Substance Use and Housing First: Results of a Randomized Trial. Addiction 2015, 110, 1605–1614. [Google Scholar] [CrossRef]
  158. Li, W.; Garland, E.L.; Howard, M.O. Therapeutic Mechanisms of Mindfulness-Oriented Recovery Enhancement for Internet Gaming Disorder: Reducing Craving and Addictive Behavior by Targeting Cognitive Processes. J. Addict. Dis. 2018, 37, 5–13. [Google Scholar] [CrossRef] [PubMed]
  159. Edwards, R.T.; McCormick-Deaton, C.; Hosanagar, A. Acute Urinary Retention Secondary to Buprenorphine Administration. Am. J. Emerg. Med. 2014, 32, e1–e2. [Google Scholar] [CrossRef] [PubMed]
  160. Nojkov, B.; Cappell, M.S. Distinctive Aspects of Peptic Ulcer Disease, Dieulafoy’s Lesion, and Mallory-Weiss Syndrome in Patients with Advanced Alcoholic Liver Disease or Cirrhosis. World J. Gastroenterol. 2016, 22, 446–466. [Google Scholar] [CrossRef] [PubMed]
  161. Kawai, S.; Yokota, T.; Onozawa, Y.; Hamauchi, S.; Fukutomi, A.; Ogawa, H.; Onoe, T.; Onitsuka, T.; Yurikusa, T.; Todaka, A.; et al. Risk Factors for Aspiration Pneumonia after Definitive Chemoradiotherapy or Bio-Radiotherapy for Locally Advanced Head and Neck Cancer: A Monocentric Case Control Study. BMC Cancer 2017, 17, 59. [Google Scholar] [CrossRef] [Green Version]
  162. Corazza, G.R.; Addolorato, G.; Biagi, F.; Caputo, F.; Castelli, E.; Stefanini, G.F.; Gasbarrini, G. Splenic Function and Alcohol Addiction. Alcohol. Clin. Exp. Res. 1997, 21, 197–200. [Google Scholar] [CrossRef]
  163. Mangla, G.; Garg, N.; Bansal, D.; Kotru, M.; Sikka, M. Peripheral Blood and Bone Marrow Findings in Chronic Alcoholics with Special Reference to Acquired Sideroblastic Anemia. Indian J. Hematol. Blood Transfus. 2020, 36, 559–564. [Google Scholar] [CrossRef]
  164. Dasarathy, J.; McCullough, A.J.; Dasarathy, S. Sarcopenia in Alcoholic Liver Disease: Clinical and Molecular Advances. Alcohol. Clin. Exp. Res. 2017, 41, 1419–1431. [Google Scholar] [CrossRef]
  165. Del Río, F.J.; Cabello, F.; Fernández, I. Influence of Substance Use on the Erectile Response in a Sample of Drug Users. Int. J. Clin. Health Psychol. 2015, 15, 37–43. [Google Scholar] [CrossRef] [Green Version]
  166. Liu, S.W.; Lien, M.H.; Fenske, N.A. The Effects of Alcohol and Drug Abuse on the Skin. Clin. Dermatol. 2010, 28, 391–399. [Google Scholar] [CrossRef]
  167. Aiempanakit, K. Crusted Scabies in a Patient with Methamphetamine Abuse. JAAD Case Rep. 2018, 4, 480–481. [Google Scholar] [CrossRef]
  168. Garcia, M.; Mulvagh, S.L.; Merz, C.N.B.; Buring, J.E.; Manson, J.E. Cardiovascular Disease in Women: Clinical Perspectives. Circ. Res. 2016, 118, 1273–1293. [Google Scholar] [CrossRef] [PubMed]
  169. Blum, K.; Schoenthaler, S.J.; Oscar-Berman, M.; Giordano, J.; Madigan, M.A.; Braverman, E.R.; Han, D. Drug Abuse Relapse Rates Linked to Level of Education: Can We Repair Hypodopaminergic-Induced Cognitive Decline with Nutrient Therapy? Phys. Sportsmed. 2014, 42, 130–145. [Google Scholar] [CrossRef] [PubMed]
  170. Caseiro, O.; Pérez-Iglesias, R.; Mata, I.; Martínez-Garcia, O.; Pelayo-Terán, J.M.; Tabares-Seisdedos, R.; Ortiz-García de la Foz, V.; Vázquez-Barquero, J.L.; Crespo-Facorro, B. Predicting Relapse after a First Episode of Non-Affective Psychosis: A Three-Year Follow-up Study. J. Psychiatr. Res. 2012, 46, 1099–1105. [Google Scholar] [CrossRef] [PubMed]
  171. Fried, L.; Modestino, E.J.; Siwicki, D.; Lott, L.; Thanos, P.K.; Baron, D.; Badgaiyan, R.D.; Ponce, J.V.; Giordano, J.; Downs, W.B.; et al. Hypodopaminergia and “Precision Behavioral Management” (PBM): It Is a Generational Family Affair. Curr. Pharm. Biotechnol. 2020, 21, 528–541. [Google Scholar] [CrossRef]
  172. Molina, B.S.G.; Pelham, W.E., Jr. Attention-Deficit/Hyperactivity Disorder and Risk of Substance Use Disorder: Developmental Considerations, Potential Pathways, and Opportunities for Research. Annu. Rev. Clin. Psychol. 2014, 10, 607–639. [Google Scholar] [CrossRef] [Green Version]
  173. Rognli, E.B.; Bramness, J.G. Understanding the Relationship between Amphetamines and Psychosis. Curr. Addict. Rep. 2015, 2, 285–292. [Google Scholar] [CrossRef] [Green Version]
  174. Cannella, L.A.; McGary, H.; Ramirez, S.H. Brain Interrupted: Early Life Traumatic Brain Injury and Addiction Vulnerability. Exp. Neurol. 2019, 317, 191–201. [Google Scholar] [CrossRef]
  175. American College of Obstetricians and Gynecologists. Opioid Use and Opioid Use Disorder in Pregnancy. Available online: https://acog.org/clinical/clinical-guidance/committee-opinion/articles/2017/08/opioid-use-and-opioid-use-disorder-in-pregnancy (accessed on 5 October 2021).
  176. McLaughlin, T.; Blum, K.; Steinberg, B.; Siwicki, D.; Campione, J.; Badgaiyan, R.D.; Braverman, E.R.; Modestino, E.J.; Gondré-Lewis, M.C.; Baron, D.; et al. Hypothesizing Las Vegas and Sutherland Springs Mass Shooters Suffer from Reward Deficiency Syndrome: “Born Bad”. J. Reward Defic. Syndr. Addict. Sci. 2017, 3, 28–31. [Google Scholar] [CrossRef]
  177. Thompson, R.G., Jr.; Wall, M.M.; Greenstein, E.; Grant, B.F.; Hasin, D.S. Substance-Use Disorders and Poverty as Prospective Predictors of First-Time Homelessness in the United States. Am. J. Public Health 2013, 103, S282–S288. [Google Scholar] [CrossRef]
  178. National Institute on Drug Abuse. Connections between Sleep and Substance Use Disorders. Available online: https://www.drugabuse.gov/about-nida/noras-blog/2020/03/connections-between-sleep-substance-use-disorders (accessed on 5 October 2021).
  179. Hazrati, L.-N.; Schwab, N. Embracing the Unknown in the Diagnosis of Traumatic Encephalopathy Syndrome. Neurology 2021, 96, 835–836. [Google Scholar] [CrossRef]
  180. Gold, M.S.; Blum, K.; Oscar-Berman, M.; Braverman, E.R. Low Dopamine Function in Attention Deficit/Hyperactivity Disorder: Should Genotyping Signify Early Diagnosis in Children? Postgrad. Med. 2014, 126, 153–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Blum, K.; Thompson, B.; Demotrovics, Z.; Femino, J.; Giordano, J.; Oscar-Berman, M.; Teitelbaum, S.; Smith, D.E.; Roy, A.K.; Agan, G.; et al. The Molecular Neurobiology of Twelve Steps Program & Fellowship: Connecting the Dots for Recovery. J. Reward Defic. Syndr. 2015, 1, 46–64. [Google Scholar] [PubMed] [Green Version]
  182. Bowen, S.; Witkiewitz, K.; Clifasefi, S.L.; Grow, J.; Chawla, N.; Hsu, S.H.; Carroll, H.A.; Harrop, E.; Collins, S.E.; Lustyk, M.K.; et al. Relative Efficacy of Mindfulness-Based Relapse Prevention, Standard Relapse Prevention, and Treatment as Usual for Substance Use Disorders: A Randomized Clinical Trial: A Randomized Clinical Trial. JAMA Psychiatry 2014, 71, 547–556. [Google Scholar] [CrossRef] [PubMed]
  183. National Institute on Drug Abuse. Treatment and Recovery. Available online: https://www.drugabuse.gov/publications/drugs-brains-behavior-science-addiction/treatment-recovery (accessed on 28 September 2021).
  184. Dakkak, M. Medications for Smoking Cessation: Guidelines from the American Thoracic Society. Am. Fam. Physician 2021, 103, 380–381. [Google Scholar]
  185. Blum, K.; Chen, A.L.C.; Chen, T.J.H.; Braverman, E.R.; Reinking, J.; Blum, S.H.; Cassel, K.; Downs, B.W.; Waite, R.L.; Williams, L.; et al. Activation Instead of Blocking Mesolimbic Dopaminergic Reward Circuitry Is a Preferred Modality in the Long Term Treatment of Reward Deficiency Syndrome (RDS): A Commentary. Theor. Biol. Med. Model. 2008, 5, 24. [Google Scholar] [CrossRef] [Green Version]
  186. Weiss, R.D.; O’malley, S.S.; Hosking, J.D.; Locastro, J.S.; Swift, R.; COMBINE Study Research Group. Do Patients with Alcohol Dependence Respond to Placebo? Results from the COMBINE Study. J. Stud. Alcohol Drugs 2008, 69, 878–884. [Google Scholar] [CrossRef]
  187. U.S. Department of Health and Human Services. (n.d.). Nimh Mental Illness. National Institute of Mental Health. Available online: https://www.nimh.nih.gov/health/statistics/mental-illness (accessed on 5 October 2021).
  188. Han, B.; Compton, W.M.; Blanco, C.; Colpe, L.J. Prevalence, Treatment, and Unmet Treatment Needs of US Adults with Mental Health and Substance Use Disorders. Health Aff. 2017, 36, 1739–1747. [Google Scholar] [CrossRef]
  189. Blum, K.; Baron, D. Opioid Substitution Therapy: Achieving Harm Reduction While Searching for a Prophylactic Solution. Curr. Pharm. Biotechnol. 2019, 20, 180–182. [Google Scholar] [CrossRef]
  190. Downs, B.W.; Blum, K.; Bagchi, D.; Kushner, S.; Bagchi, M.; Galvin, J.M.; Lewis, M.; Siwicki, D.; Brewer, R.; Boyett, B.; et al. Molecular Neuro-Biological and Systemic Health Benefits of Achieving Dopamine Homeostasis in the Face of a Catastrophic Pandemic (COVID- 19): A Mechanistic Exploration. J. Syst. Integr. Neurosci. 2020, 7. [Google Scholar] [CrossRef]
  191. McDonald, R.D.; Tofighi, B.; Laska, E.; Goldfeld, K.; Bonilla, W.; Flannery, M.; Santana-Correa, N.; Johnson, C.W.; Leibowitz, N.; Rotrosen, J.; et al. Extended-Release Naltrexone Opioid Treatment at Jail Reentry (XOR). Contemp. Clin. Trials 2016, 49, 57–64. [Google Scholar] [CrossRef] [Green Version]
  192. Swift, R.; Oslin, D.W.; Alexander, M.; Forman, R. Adherence Monitoring in Naltrexone Pharmacotherapy Trials: A Systematic Review. J. Stud. Alcohol Drugs 2011, 72, 1012–1018. [Google Scholar] [CrossRef] [PubMed]
  193. Williams, A.R.; Barbieri, V.; Mishlen, K.; Levin, F.R.; Nunes, E.V.; Mariani, J.J.; Bisaga, A. Long-Term Follow-up Study of Community-Based Patients Receiving XR-NTX for Opioid Use Disorders. Am. J. Addict. 2017, 26, 319–325. [Google Scholar] [CrossRef] [PubMed]
  194. Srivastava, A.B.; Gold, M.S. Beyond Supply: How We Must Tackle the Opioid Epidemic. Mayo Clin. Proc. 2018, 93, 269–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Blum, K.; Lott, L.; Baron, D.; Smith, D.E.; Badgaiyan, R.D.; Gold, M.S. Improving Naltrexone Compliance and Outcomes with Putative Pro- Dopamine Regulator KB220, Compared to Treatment as Usual. J. Syst. Integr. Neurosci. 2020, 7. [Google Scholar] [CrossRef] [PubMed]
  196. Blum, K.; Modestino, E.J.; Gondre-Lewis, M.C.; Baron, D.; Steinberg, B.; Thanos P, K.; Downs, W.B.; Davis, S.; Lisa, L.; Eric, B.R.; et al. Pro-Dopamine Regulator (KB220) A Fifty Year Sojourn to Combat Reward Deficiency Syndrome (RDS): Evidence Based Bibliography (Annotated). CPQ Neurol Psychol 2018, 1. [Google Scholar]
  197. Gratwicke, J.; Oswal, A.; Akram, H.; Jahanshahi, M.; Hariz, M.; Zrinzo, L.; Foltynie, T.; Litvak, V. Resting State Activity and Connectivity of the Nucleus Basalis of Meynert and Globus Pallidus in Lewy Body Dementia and Parkinson’s Disease Dementia. Neuroimage 2020, 221, 117184. [Google Scholar] [CrossRef]
  198. Cadet, J.L.; Bisagno, V. Neuropsychological Consequences of Chronic Drug Use: Relevance to Treatment Approaches. Front. Psychiatry 2015, 6, 189. [Google Scholar] [CrossRef]
  199. Bastos Leite, A.J.; van der Flier, W.M.; van Straaten, E.C.W.; Scheltens, P.; Barkhof, F. Infratentorial Abnormalities in Vascular Dementia. Stroke 2006, 37, 105–110. [Google Scholar] [CrossRef] [Green Version]
  200. Frings, L.; Klöppel, S.; Teipel, S.; Peters, O.; Frölich, L.; Pantel, J.; Schröder, J.; Gertz, H.-J.; Arlt, S.; Heuser, I.; et al. Left Anterior Temporal Lobe Sustains Naming in Alzheimer’s Dementia and Mild Cognitive Impairment. Curr. Alzheimer Res. 2011, 8, 893–901. [Google Scholar] [CrossRef]
  201. Ziabreva, I.; Ballard, C.G.; Aarsland, D.; Larsen, J.-P.; McKeith, I.G.; Perry, R.H.; Perry, E.K. Lewy Body Disease: Thalamic Cholinergic Activity Related to Dementia and Parkinsonism. Neurobiol. Aging 2006, 27, 433–438. [Google Scholar] [CrossRef]
  202. Akanuma, K.; Meguro, K.; Meguro, M.; Sasaki, E.; Chiba, K.; Ishii, H.; Tanaka, N. Improved Social Interaction and Increased Anterior Cingulate Metabolism after Group Reminiscence with Reality Orientation Approach for Vascular Dementia. Psychiatry Res. 2011, 192, 183–187. [Google Scholar] [CrossRef] [PubMed]
  203. Barnes, J.; Godbolt, A.K.; Frost, C.; Boyes, R.G.; Jones, B.F.; Scahill, R.I.; Rossor, M.N.; Fox, N.C. Atrophy Rates of the Cingulate Gyrus and Hippocampus in AD and FTLD. Neurobiol. Aging 2007, 28, 20–28. [Google Scholar] [CrossRef] [PubMed]
  204. Bozzali, M.; D’Amelio, M.; Serra, L. Ventral Tegmental Area Disruption in Alzheimer’s Disease. Aging 2019, 11, 1325–1326. [Google Scholar] [CrossRef] [PubMed]
  205. Brambati, S.M.; Renda, N.C.; Rankin, K.P.; Rosen, H.J.; Seeley, W.W.; Ashburner, J.; Weiner, M.W.; Miller, B.L.; Gorno-Tempini, M.L. A Tensor Based Morphometry Study of Longitudinal Gray Matter Contraction in FTD. Neuroimage 2007, 35, 998–1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Cousins, D.A.; Burton, E.J.; Burn, D.; Gholkar, A.; McKeith, I.G.; O’Brien, J.T. Atrophy of the Putamen in Dementia with Lewy Bodies but Not Alzheimer’s Disease: An MRI Study. Neurology 2003, 61, 1191–1195. [Google Scholar] [CrossRef] [PubMed]
  207. Freedman, M.; Binns, M.A.; Black, S.E.; Levine, B.; Miller, B.L.; Ramirez, J.; Szilagyi, G.M.; Scott, C.J.M.; McNeely, A.A.; Stuss, D.T. Object Alternation: A Novel Probe of Medial Frontal Function in Frontotemporal Dementia. Alzheimer Dis. Assoc. Disord. 2013, 27, 316–323. [Google Scholar] [CrossRef] [Green Version]
  208. Guo, C.C.; Tan, R.; Hodges, J.R.; Hu, X.; Sami, S.; Hornberger, M. Network-Selective Vulnerability of the Human Cerebellum to Alzheimer’s Disease and Frontotemporal Dementia. Brain 2016, 139, 1527–1538. [Google Scholar] [CrossRef]
  209. Kawakami, I.; Hasegawa, M.; Arai, T.; Ikeda, K.; Oshima, K.; Niizato, K.; Aoki, N.; Omi, K.; Higashi, S.; Hosokawa, M.; et al. Tau Accumulation in the Nucleus Accumbens in Tangle-Predominant Dementia. Acta Neuropathol. Commun. 2014, 2, 40. [Google Scholar] [CrossRef] [Green Version]
  210. Lee, Y.H.; Bak, Y.; Park, C.-H.; Chung, S.J.; Yoo, H.S.; Baik, K.; Jung, J.H.; Sohn, Y.H.; Shin, N.-Y.; Lee, P.H. Patterns of Olfactory Functional Networks in Parkinson’s Disease Dementia and Alzheimer’s Dementia. Neurobiol. Aging 2020, 89, 63–70. [Google Scholar] [CrossRef]
  211. Lee, Y.-G.; Jeon, S.; Yoo, H.S.; Chung, S.J.; Lee, S.-K.; Lee, P.H.; Sohn, Y.H.; Yun, M.; Evans, A.C.; Ye, B.S. Amyloid-β-Related and Unrelated Cortical Thinning in Dementia with Lewy Bodies. Neurobiol. Aging 2018, 72, 32–39. [Google Scholar] [CrossRef]
  212. Liu, Z.; Wei, W.; Bai, L.; Dai, R.; You, Y.; Chen, S.; Tian, J. Exploring the Patterns of Acupuncture on Mild Cognitive Impairment Patients Using Regional Homogeneity. PLoS ONE 2014, 9, e99335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Maetzler, W.; Reimold, M.; Liepelt, I.; Solbach, C.; Leyhe, T.; Schweitzer, K.; Eschweiler, G.W.; Mittelbronn, M.; Gaenslen, A.; Uebele, M.; et al. [11C] PIB Binding in Parkinson’s Disease Dementia. Neuroimage 2008, 39, 1027–1033. [Google Scholar] [CrossRef] [PubMed]
  214. Oh, H.; Gosnell, S.; Nguyen, T.; Tran, T.; Kosten, T.R.; Salas, R. Cingulate Cortex Structural Alterations in Substance Use Disorder Psychiatric Inpatients. Am. J. Addict. 2021, 30, 72–79. [Google Scholar] [CrossRef]
  215. Mega, M.S.; Lee, L.; Dinov, I.D.; Mishkin, F.; Toga, A.W.; Cummings, J.L. Cerebral Correlates of Psychotic Symptoms in Alzheimer’s Disease. J. Neurol. Neurosurg. Psychiatry 2000, 69, 167–171. [Google Scholar] [CrossRef] [PubMed]
  216. Moretti, R.; Torre, P.; Antonello, R.M.; Cattaruzza, T.; Cazzato, G.; Bava, A. Frontal Lobe Dementia and Subcortical Vascular Dementia: A Neuropsychological Comparison. Psychol. Rep. 2005, 96, 141–151. [Google Scholar] [CrossRef] [PubMed]
  217. Prosser, A.M.J.; Tossici-Bolt, L.; Kipps, C.M. Occipital Lobe and Posterior Cingulate Perfusion in the Prediction of Dementia with Lewy Body Pathology in a Clinical Sample. Nucl. Med. Commun. 2017, 38, 1029–1035. [Google Scholar] [CrossRef]
  218. Pucci, E.; Belardinelli, N.; Regnicolo, L.; Nolfe, G.; Signorino, M.; Salvolini, U.; Angeleri, F. Hippocampus and Parahippocampal Gyrus Linear Measurements Based on Magnetic Resonance in Alzheimer’s Disease. Eur. Neurol. 1998, 39, 16–25. [Google Scholar] [CrossRef]
  219. Ross, A.J.; Sachdev, P.S.; Wen, W.; Valenzuela, M.J.; Brodaty, H. 1H MRS in Stroke Patients with and without Cognitive Impairment. Neurobiol. Aging 2005, 26, 873–882. [Google Scholar] [CrossRef]
  220. Schuff, N.; Capizzano, A.A.; Du, A.T.; Amend, D.L.; O’Neill, J.; Norman, D.; Jagust, W.J.; Chui, H.C.; Kramer, J.H.; Reed, B.R.; et al. Different Patterns of N-Acetylaspartate Loss in Subcortical Ischemic Vascular Dementia and AD. Neurology 2003, 61, 358–364. [Google Scholar] [CrossRef] [Green Version]
  221. Schwab, S.; Afyouni, S.; Chen, Y.; Han, Z.; Guo, Q.; Dierks, T.; Wahlund, L.-O.; Grieder, M. Functional Connectivity Alterations of the Temporal Lobe and Hippocampus in Semantic Dementia and Alzheimer’s Disease. J. Alzheimers. Dis. 2020, 76, 1461–1475. [Google Scholar] [CrossRef]
  222. Sung, Y.-H.; Park, K.-H.; Lee, Y.-B.; Park, H.-M.; Shin, D.J.; Park, J.-S.; Oh, M.-S.; Ma, H.-I.; Yu, K.-H.; Kang, S.-Y.; et al. Midbrain Atrophy in Subcortical Ischemic Vascular Dementia. J. Neurol. 2009, 256, 1997–2002. [Google Scholar] [CrossRef] [PubMed]
  223. van Marle, H.J.F.; Tendolkar, I.; Urner, M.; Verkes, R.J.; Fernández, G.; van Wingen, G. Subchronic Duloxetine Administration Alters the Extended Amygdala Circuitry in Healthy Individuals. Neuroimage 2011, 55, 825–831. [Google Scholar] [CrossRef] [PubMed]
  224. Vitanova, K.S.; Stringer, K.M.; Benitez, D.P.; Brenton, J.; Cummings, D.M. Dementia Associated with Disorders of the Basal Ganglia. J. Neurosci. Res. 2019, 97, 1728–1741. [Google Scholar] [CrossRef] [PubMed]
  225. Tak, K.; Lee, S.; Choi, E.; Suh, S.W.; Oh, D.J.; Moon, W.; Kim, H.S.; Byun, S.; Bae, J.B.; Han, J.W.; et al. Magnetic Resonance Imaging Texture of Medial Pulvinar in Dementia with Lewy Bodies. Dement. Geriatr. Cogn. Disord. 2020, 49, 8–15. [Google Scholar] [CrossRef]
  226. Burgmans, S.; van Boxtel, M.P.J.; Smeets, F.; Vuurman, E.F.P.M.; Gronenschild, E.H.B.M.; Verhey, F.R.J.; Uylings, H.B.M.; Jolles, J. Prefrontal Cortex Atrophy Predicts Dementia over a Six-Year Period. Neurobiol. Aging 2009, 30, 1413–1419. [Google Scholar] [CrossRef] [Green Version]
  227. Brickman, A.M.; Meier, I.B.; Korgaonkar, M.S.; Provenzano, F.A.; Grieve, S.M.; Siedlecki, K.L.; Wasserman, B.T.; Williams, L.M.; Zimmerman, M.E. Testing the White Matter Retrogenesis Hypothesis of Cognitive Aging. Neurobiol. Aging 2012, 33, 1699–1715. [Google Scholar] [CrossRef] [Green Version]
  228. Tsai, S.-T.; Liew, H.-K.; Li, H.-M.; Lin, S.-Z.; Chen, S.-Y. Harnessing Neurogenesis and Neuroplasticity with Stem Cell Treatment for Addictive Disorders. Cell Transplant. 2019, 28, 1127–1131. [Google Scholar] [CrossRef] [Green Version]
  229. Chambers, R.A. Adult Hippocampal Neurogenesis in the Pathogenesis of Addiction and Dual Diagnosis Disorders. Drug Alcohol Depend. 2013, 130, 1–12. [Google Scholar] [CrossRef] [Green Version]
  230. Canales, J.J. Deficient Plasticity in the Hippocampus and the Spiral of Addiction: Focus on Adult Neurogenesis. Curr. Top. Behav. Neurosci. 2013, 15, 293–312. [Google Scholar]
  231. Crews, F.T.; Nixon, K. Alcohol, Neural Stem Cells, and Adult Neurogenesis. Alcohol Res. Health 2003, 27, 197–204. [Google Scholar]
  232. Nixon, K. Alcohol and Adult Neurogenesis: Roles in Neurodegeneration and Recovery in Chronic Alcoholism. Hippocampus 2006, 16, 287–295. [Google Scholar] [CrossRef] [PubMed]
  233. Schlagal, C.R.; Wu, P. Alcohol and Cocaine Combined Substance Use on Adult Hypothalamic Neural Stem Cells and Neurogenesis. Brain Plast. 2020, 6, 41–46. [Google Scholar] [CrossRef] [PubMed]
  234. Famitafreshi, H.; Karimian, M.; Fatima, S. Synergistic Effects of Social Isolation and Morphine Addiction on Reduced Neurogenesis and BDNF Levels and the Resultant Deficits in Cognition and Emotional State in Male Rats. Curr. Mol. Pharmacol. 2016, 9, 337–347. [Google Scholar] [CrossRef] [PubMed]
  235. Li, Y.; Shen, M.; Stockton, M.E.; Zhao, X. Hippocampal Deficits in Neurodevelopmental Disorders. Neurobiol. Learn. Mem. 2019, 165, 106945. [Google Scholar] [CrossRef]
  236. Gualtieri, C.T.; Johnson, L.G. Reliability and Validity of a Computerized Neurocognitive Test Battery, CNS Vital Signs. Arch. Clin. Neuropsychol. 2006, 21, 623–643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Campman, C.; van Ranst, D.; Meijer, J.W.; Sitskoorn, M. Computerized Screening for Cognitive Impairment in Patients with COPD. Int. J. Chron. Obstruct. Pulmon. Dis. 2017, 12, 3075–3083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Olofsen, E.; Noppers, I.; Niesters, M.; Kharasch, E.; Aarts, L.; Sarton, E.; Dahan, A. Estimation of the Contribution of Norketamine to Ketamine-Induced Acute Pain Relief and Neurocognitive Impairment in Healthy Volunteers. Anesthesiology 2012, 117, 353–364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Meador, K.J.; Seliger, J.; Boyd, A.; Razavi, B.; Falco-Walter, J.; Le, S.; Loring, D.W. Comparative Neuropsychological Effects of Carbamazepine and Eslicarbazepine Acetate. Epilepsy Behav. 2019, 94, 151–157. [Google Scholar] [CrossRef]
  240. Littleton, A.C.; Register-Mihalik, J.K.; Guskiewicz, K.M. Test-Retest Reliability of a Computerized Concussion Test: CNS Vital Signs: CNS Vital Signs. Sports Health 2015, 7, 443–447. [Google Scholar] [CrossRef] [Green Version]
  241. Brooks, B.L.; Plourde, V.; Fay-McClymont, T.B.; MacAllister, W.S.; Sherman, E.M.S. Factor Structure of the CNS Vital Signs Computerized Cognitive Battery in Youth with Neurological Diagnoses. Child Neuropsychol. 2019, 25, 980–991. [Google Scholar] [CrossRef]
  242. Bojar, I.; Wojcik-Fatla, A.; Owoc, A.; Lewinski, A. Polymorphisms of Apolipoprotein E Gene and Cognitive Functions of Postmenopausal Women, Measured by Battery of Computer Tests—Central Nervous System Vital Signs. Neuro Endocrinol. Lett. 2012, 33, 385–392. [Google Scholar] [PubMed]
  243. Van de Glind, G.; Brynte, C.; Skutle, A.; Kaye, S.; Konstenius, M.; Levin, F.; Mathys, F.; Demetrovics, Z.; Moggi, F.; Ramos-Quiroga, J.A.; et al. The International Collaboration on ADHD and Substance Abuse (ICASA): Mission, Results, and Future Activities. Eur. Addict. Res. 2020, 26, 173–178. [Google Scholar] [CrossRef] [PubMed]
  244. Assayag, N.; Berger, I.; Parush, S.; Mell, H.; Bar-Shalita, T. Attention-Deficit/Hyperactivity Disorder Symptoms, Sensation-Seeking, and Sensory Modulation Dysfunction in Substance Use Disorder: A Cross Sectional Two-Group Comparative Study. Int. J. Environ. Res. Public Health 2022, 19, 2541. [Google Scholar] [CrossRef] [PubMed]
  245. Braverman, E.R.; Chen, T.J.H.; Schoolfield, J.; Martinez-Pons, M.; Arcuri, V.; Varshavskiy, M.; Gordon, C.A.; Mengucci, J.; Blum, S.H.; Meshkin, B.; et al. Delayed P300 Latency Correlates with Abnormal Test of Variables of Attention (TOVA) in Adults and Predicts Early Cognitive Decline in a Clinical Setting. Adv. Ther. 2006, 23, 582–600. [Google Scholar] [CrossRef]
  246. Braverman, E.R.; Chen, A.L.-C.; Chen, T.J.H.; Schoolfield, J.D.; Notaro, A.; Braverman, D.; Kerner, M.; Blum, S.H.; Arcuri, V.; Varshavskiy, M.; et al. Test of Variables of Attention (TOVA) as a Predictor of Early Attention Complaints, an Antecedent to Dementia. Neuropsychiatr. Dis. Treat. 2010, 6, 681–690. [Google Scholar]
  247. Bodkyn, C.N.; Holroyd, C.B. Neural Mechanisms of Affective Instability and Cognitive Control in Substance Use. Int. J. Psychophysiol. 2019, 146, 1–19. [Google Scholar] [CrossRef]
  248. Gruber, R.; Grizenko, N.; Schwartz, G.; Bellingham, J.; Guzman, R.; Joober, R. Performance on the Continuous Performance Test in Children with ADHD Is Associated with Sleep Efficiency. Sleep 2007, 30, 1003–1009. [Google Scholar] [CrossRef] [Green Version]
  249. Grimm, O.; van Rooij, D.; Tshagharyan, A.; Yildiz, D.; Leonards, J.; Elgohary, A.; Buitelaar, J.; Reif, A. Effects of Comorbid Disorders on Reward Processing and Connectivity in Adults with ADHD. Transl. Psychiatry 2021, 11, 636. [Google Scholar] [CrossRef]
  250. Wiedmann, M.; Atzendorf, J.; Basedow, L.A.; Roessner, V.; Golub, Y.; Kuitunen-Paul, S. Substanzkonsum, Störungen durch Substanzkonsum und begleitende psychische Störungen bei Jugendlichen: Zahlen aus einer Spezialambulanz für Suchterkrankungen [Substance Use, Resulting Disorders, and Collateral Mental Disorders Among Adolescents in a Special Outpatient Institutions for Addictions]. Z. Kinder Jugendpsychiatr. Psychother. 2021, 50, 105–119. [Google Scholar] [CrossRef]
  251. Jackson, N.J.; Isen, J.D.; Khoddam, R.; Irons, D.; Tuvblad, C.; Iacono, W.G.; McGue, M.; Raine, A.; Baker, L.A. Impact of Adolescent Marijuana Use on Intelligence: Results from Two Longitudinal Twin Studies. Proc. Natl. Acad. Sci. USA 2016, 113, E500-8. [Google Scholar] [CrossRef] [Green Version]
  252. Ball, S.A.; Nich, C.; Rounsaville, B.J.; Eagan, D.; Carroll, K.M. Millon Clinical Multiaxial Inventory-III Subtypes of Opioid Dependence: Validity and Matching to Behavioral Therapies. J. Consult. Clin. Psychol. 2004, 72, 698–711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Strack, S.; Millon, T. Contributions to the Dimensional Assessment of Personality Disorders Using Millon’s Model and the Millon Clinical Multiaxial Inventory (MCMI9-III). J. Pers. Assess. 2007, 89, 56–69. [Google Scholar] [CrossRef] [PubMed]
  254. Millon, T. Millon Clinical Multiaxial Inventory: I & II. J. Couns. Dev. 1992, 70, 421–426. [Google Scholar]
  255. Choca, J.P.; Grossman, S.D. Evolution of the Millon Clinical Multiaxial Inventory. J. Pers. Assess. 2015, 97, 541–549. [Google Scholar] [CrossRef] [PubMed]
  256. Millon, C.; Millon, T. Q-Global Millon Clinical Multiaxial Inventory III Corrections Interpretive Report (MCMI-III), 3rd ed.; Psychological Corporation: London, UK, 2019. [Google Scholar]
  257. Braverman, E.R.; Blum, K. Substance Use Disorder Exacerbates Brain Electrophysiological Abnormalities in a Psychiatrically-Ill Population. Clin. Electroencephalogr. 1996, 27, 5–27. [Google Scholar] [CrossRef] [PubMed]
  258. Krepel, N.; Egtberts, T.; Sack, A.T.; Heinrich, H.; Ryan, M.; Arns, M. A Multicenter Effectiveness Trial of QEEG-Informed Neurofeedback in ADHD: Replication and Treatment Prediction. NeuroImage Clin. 2020, 28, 102399. [Google Scholar] [CrossRef] [PubMed]
  259. National Institute on Drug Abuse. Drug Misuse and Addiction. Available online: https://www.drugabuse.gov/publications/drugs-brains-behavior-science-addiction/drug-misuse-addiction (accessed on 28 September 2021).
  260. Blum, K.; Chen, T.J.H.; Morse, S.; Giordano, J.; Chen, A.L.C.; Thompson, J.; Allen, C.; Smolen, A.; Lubar, J.; Stice, E.; et al. Overcoming QEEG Abnormalities and Reward Gene Deficits during Protracted Abstinence in Male Psychostimulant and Polydrug Abusers Utilizing Putative Dopamine D2 Agonist Therapy: Part 2. Postgrad. Med. 2010, 122, 214–226. [Google Scholar] [CrossRef] [Green Version]
  261. Burns, J.A.; Kroll, D.S.; Feldman, D.E.; Kure Liu, C.; Manza, P.; Wiers, C.E.; Volkow, N.D.; Wang, G.-J. Molecular Imaging of Opioid and Dopamine Systems: Insights into the Pharmacogenetics of Opioid Use Disorders. Front. Psychiatry 2019, 10, 626. [Google Scholar] [CrossRef]
  262. Boileau, I.; Payer, D.; Rusjan, P.M.; Houle, S.; Tong, J.; McCluskey, T.; Wilson, A.A.; Kish, S.J. Heightened Dopaminergic Response to Amphetamine at the D3 Dopamine Receptor in Methamphetamine Users. Neuropsychopharmacology 2016, 41, 2994–3002. [Google Scholar] [CrossRef] [Green Version]
  263. Wang, Y.; Yan, K.-J.; Fan, C.-X.; Luo, X.-N.; Zhou, Y. Altered Functional Connectivity of the Nucleus Accumbens Subdivisions in Amphetamine-Type Stimulant Abusers: A Resting-State FMRI Study. BMC Neurosci. 2019, 20, 66. [Google Scholar] [CrossRef]
  264. Sokhadze, T.M.; Cannon, R.L.; Trudeau, D.L. EEG Biofeedback as a Treatment for Substance Use Disorders: Review, Rating of Efficacy, and Recommendations for Further Research. Appl. Psychophysiol. Biofeedback 2008, 33, 1–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Feldstein Ewing, S.W.; Filbey, F.M.; Chandler, L.D.; Hutchison, K.E. Exploring the Relationship between Depressive and Anxiety Symptoms and Neuronal Response to Alcohol Cues. Alcohol. Clin. Exp. Res. 2010, 34, 396–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Van Dinteren, R.; Arns, M.; Jongsma, M.L.A.; Kessels, R.P.C. P300 Development across the Lifespan: A Systematic Review and Meta-Analysis. PLoS ONE 2014, 9, e87347. [Google Scholar] [CrossRef] [PubMed]
  267. Braverman, E.R.; Han, D.; Oscar-Berman, M.; Karikh, T.; Truesdell, C.; Dushaj, K.; Kreuk, F.; Li, M.; Stratton, D.; Blum, K. Menopause Analytical Hormonal Correlate Outcome Study (MAHCOS) and the Association to Brain Electrophysiology (P300) in a Clinical Setting. PLoS ONE 2014, 9, e105048. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  268. Braverman, E.R.; Blum, K. P300 (Latency) Event-Related Potential: An Accurate Predictor of Memory Impairment. Clin. Electroencephalogr. 2003, 34, 124–139. [Google Scholar] [CrossRef] [PubMed]
  269. Braverman, E. Use of Growth Hormone in Elderly Individuals. JAMA 2003, 290, 462. [Google Scholar]
  270. Braverman, E.R.; Chen, T.J.H.; Chen, A.L.C.; Kerner, M.M.; Tung, H.; Waite, R.L.; Schoolfield, J.; Blum, K. Preliminary Investigation of Plasma Levels of Sex Hormones and Human Growth Factor(s), and P300 Latency as Correlates to Cognitive Decline as a Function of Gender. BMC Res. Notes 2009, 2, 126. [Google Scholar] [CrossRef] [Green Version]
  271. Braverman, E.R.; Chen, T.J.H.; Prihoda, T.J.; Sonntag, W.; Meshkin, B.; Downs, B.W.; Mengucci, J.F.; Blum, S.H.; Notaro, A.; Arcuri, V.; et al. Plasma Growth Hormones, P300 Event-Related Potential and Test of Variables of Attention (TOVA) Are Important Neuroendocrinological Predictors of Early Cognitive Decline in a Clinical Setting: Evidence Supported by Structural Equation Modeling (SEM) Parameter Estimates. Age 2007, 29, 55–67. [Google Scholar]
  272. Mason, B.L.; Van Enkevort, E.; Filbey, F.; Marx, C.E.; Park, J.; Nakamura, A.; Sunderajan, P.; Brown, E.S. Neurosteroid Levels in Patients with Bipolar Disorder and a History of Cannabis Use Disorders. J. Clin. Psychopharmacol. 2017, 37, 684–688. [Google Scholar] [CrossRef]
  273. Lorberboym, M.; Gilad, R.; Gorin, V.; Sadeh, M.; Lampl, Y. Late Whiplash Syndrome: Correlation of Brain SPECT with Neuropsychological Tests and P300 Event-Related Potential. J. Trauma 2002, 52, 521–526. [Google Scholar] [CrossRef]
  274. Braverman, E.R.; Blum, K.; Hussman, K.L.; Han, D.; Dushaj, K.; Li, M.; Marin, G.; Badgaiyan, R.D.; Smayda, R.; Gold, M.S. Evoked Potentials and Memory/Cognition Tests Validate Brain Atrophy as Measured by 3T MRI (NeuroQuant) in Cognitively Impaired Patients. PLoS ONE 2015, 10, e0133609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Filbey, F.M.; McQueeny, T.; DeWitt, S.J.; Mishra, V. Preliminary Findings Demonstrating Latent Effects of Early Adolescent Marijuana Use Onset on Cortical Architecture. Dev. Cogn. Neurosci. 2015, 16, 16–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  276. Thayer, R.E.; Hansen, N.S.; Prashad, S.; Karoly, H.C.; Filbey, F.M.; Bryan, A.D.; Feldstein Ewing, S.W. Recent Tobacco Use Has Widespread Associations with Adolescent White Matter Microstructure. Addict. Behav. 2020, 101, 106152. [Google Scholar] [CrossRef] [PubMed]
  277. Braverman, E.R.; Blum, K.; Damle, U.J.; Kerner, M.; Dushaj, K.; Oscar-Berman, M. Evoked Potentials and Neuropsychological Tests Validate Positron Emission Topography (PET) Brain Metabolism in Cognitively Impaired Patients. PLoS ONE 2013, 8, e55398. [Google Scholar] [CrossRef] [Green Version]
  278. Milivojevic, V.; Sinha, R. Central and Peripheral Biomarkers of Stress Response for Addiction Risk and Relapse Vulnerability. Trends Mol. Med. 2018, 24, 173–186. [Google Scholar] [CrossRef]
  279. Quinlan, E.B.; Banaschewski, T.; Barker, G.J.; Bokde, A.L.W.; Bromberg, U.; Büchel, C.; Desrivières, S.; Flor, H.; Frouin, V.; Garavan, H.; et al. Identifying Biological Markers for Improved Precision Medicine in Psychiatry. Mol. Psychiatry 2020, 25, 243–253. [Google Scholar] [CrossRef]
  280. Lesho, E.; Gey, D.; Forrester, G.; Michaud, E.; Emmons, E.; Huycke, E. The Low Impact of Screening Electrocardiograms in Healthy Individuals: A Prospective Study and Review of the Literature. Mil. Med. 2003, 168, 15–18. [Google Scholar] [CrossRef]
  281. Anderson, J.B.; Grenier, M.; Edwards, N.M.; Madsen, N.L.; Czosek, R.J.; Spar, D.S.; Barnes, A.; Pratt, J.; King, E.; Knilans, T.K. Usefulness of Combined History, Physical Examination, Electrocardiogram, and Limited Echocardiogram in Screening Adolescent Athletes for Risk for Sudden Cardiac Death. Am. J. Cardiol. 2014, 114, 1763–1767. [Google Scholar] [CrossRef]
  282. Muratova, V.N.; Islam, S.S.; Demerath, E.W.; Minor, V.E.; Neal, W.A. Cholesterol Screening among Children and Their Parents. Prev. Med. 2001, 33, 1–6. [Google Scholar] [CrossRef]
  283. Schoenthaler, S.J.; Blum, K.; Braverman, E.R.; Giordano, J.; Thompson, B.; Oscar-Berman, M.; Badgaiyan, R.D.; Madigan, M.A.; Dushaj, K.; Li, M.; et al. NIDA-Drug Addiction Treatment Outcome Study (DATOS) Relapse as a Function of Spirituality/Religiosity. J. Reward Defic. Syndr. 2015, 1, 36–45. [Google Scholar] [CrossRef] [Green Version]
  284. Blum, K.; Thompson, B.; Oscar-Berman, M.; Giordano, J.; Braverman, E.; Femino, J.; Barh, D.; Downs, W.; Smpatico, T.; Schoenthaler, S. Genospirituality: Our Beliefs, Our Genomes, and Addictions. J. Addict. Res. Ther. 2013, 5, 162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. Ulaş, E.; Ekşi, H. Inclusion of Family Therapy in Rehabilitation Program of Substance Abuse and Its Efficacious Implementation. Fam. J. Alex. Va 2019, 27, 443–451. [Google Scholar] [CrossRef]
  286. Roy, A.K.; Bowirrat, A.; Smith, D.E.; Braverman, E.R.; Jalali, R.; Badgaiyan, R.D.; Baron, D.; Gomez, L.L.; Barh, D.; Blum, K. Neurobiology and Spirituality in Addiction Recovery. Acta Sci. Neurol. 2021, 4, 64–71. [Google Scholar] [PubMed]
  287. Hallgren, K.A.; Cohn, E.B.; Ries, R.K.; Atkins, D.C. Delivering Remote Measurement-Based Care in Community Addiction Treatment: Engagement and Usability over a 6-Month Clinical Pilot. Front. Psychiatry 2022, 13, 840409. [Google Scholar] [CrossRef]
  288. Frodl-Bauch, T.; Bottlender, R.; Hegerl, U. Neurochemical Substrates and Neuroanatomical Generators of the Event-Related P300. Available online: https://epub.ub.uni-muenchen.de/16552/1/10_1159_000026603.pdf (accessed on 2 October 2021).
  289. Wang, G.Y.; Kydd, R.; Russell, B.R. Resting EEG and ERPs Findings in Methadone-Substituted Opiate Users: A Review. Acta Neurol. Belg. 2015, 115, 539–546. [Google Scholar] [CrossRef]
  290. Singh, S.M.; Basu, D. The P300 Event-Related Potential and Its Possible Role as an Endophenotype for Studying Substance Use Disorders: A Review. Addict. Biol. 2009, 14, 298–309. [Google Scholar] [CrossRef]
  291. Campanella, S.; Pogarell, O.; Boutros, N. Event-Related Potentials in Substance Use Disorders: A Narrative Review Based on Articles from 1984 to 2012: A Narrative Review Based on Articles from 1984 to 2012. Clin. EEG Neurosci. 2014, 45, 67–76. [Google Scholar] [CrossRef]
  292. Vassena, E.; Silvetti, M.; Boehler, C.N.; Achten, E.; Fias, W.; Verguts, T. Overlapping Neural Systems Represent Cognitive Effort and Reward Anticipation. PLoS ONE 2014, 9, e91008. [Google Scholar] [CrossRef]
  293. Gondré-Lewis, M.C.; Bassey, R.; Blum, K. Pre-Clinical Models of Reward Deficiency Syndrome: A Behavioral Octopus. Neurosci. Biobehav. Rev. 2020, 115, 164–188. [Google Scholar] [CrossRef]
  294. Blum, K.; Thanos, P.K.; Badgaiyan, R.D.; Febo, M.; Oscar-Berman, M.; Fratantonio, J.; Demotrovics, Z.; Gold, M.S. Neurogenetics and Gene Therapy for Reward Deficiency Syndrome: Are We Going to the Promised Land? Expert Opin. Biol. Ther. 2015, 15, 973–985. [Google Scholar] [CrossRef]
  295. Blum, K.; Bowirrat, A.; Gondre Lewis, M.C.; Simpatico, T.A.; Ceccanti, M.; Steinberg, B.; Modestino, E.J.; Thanos, P.K.; Baron, D.; McLaughlin, T.; et al. Exploration of Epigenetic State Hyperdopaminergia (Surfeit) and Genetic Trait Hypodopaminergia (Deficit) during Adolescent Brain Development. Curr. Psychopharmacol. 2021, 10, 181–196. [Google Scholar] [CrossRef] [PubMed]
  296. Blum, K. Coupling Genetic Addiction Risk Score (GARS) and pro Dopamine Regulation (KB220) to Combat Substance Use Disorder (SUD). Glob. J. Addict. Rehabil. Med. 2017, 1, 555556. [Google Scholar] [CrossRef] [PubMed]
  297. Blum, K.; Oscar-Berman, M.; Dinubile, N.; Giordano, J.; Braverman, E.R.; Truesdell, C.E.; Barh, D.; Badgaiyan, R. Coupling Genetic Addiction Risk Score (GARS) with Electrotherapy: Fighting Iatrogenic Opioid Dependence. J. Addict. Res. Ther. 2013, 4, 1000163. [Google Scholar] [PubMed] [Green Version]
  298. Li, Y.; Simmler, L.D.; Van Zessen, R.; Flakowski, J.; Wan, J.-X.; Deng, F.; Li, Y.-L.; Nautiyal, K.M.; Pascoli, V.; Lüscher, C. Synaptic Mechanism Underlying Serotonin Modulation of Transition to Cocaine Addiction. Science 2021, 373, 1252–1256. [Google Scholar] [CrossRef]
  299. Volkow, N.D.; Koob, G.F.; McLellan, A.T. Neurobiologic Advances from the Brain Disease Model of Addiction. N. Engl. J. Med. 2016, 374, 363–371. [Google Scholar] [CrossRef]
  300. Nkansah-Amankra, S.; Minelli, M. “Gateway Hypothesis” and Early Drug Use: Additional Findings from Tracking a Population-Based Sample of Adolescents to Adulthood. Prev. Med. Rep. 2016, 4, 134–141. [Google Scholar] [CrossRef] [Green Version]
  301. Otten, R.; Mun, C.J.; Dishion, T.J. The Social Exigencies of the Gateway Progression to the Use of Illicit Drugs from Adolescence into Adulthood. Addict. Behav. 2017, 73, 144–150. [Google Scholar] [CrossRef]
  302. Colon-Perez, L.M.; Tran, K.; Thompson, K.; Pace, M.C.; Blum, K.; Goldberger, B.A.; Gold, M.S.; Bruijnzeel, A.W.; Setlow, B.; Febo, M. The Psychoactive Designer Drug and Bath Salt Constituent MDPV Causes Widespread Disruption of Brain Functional Connectivity. Neuropsychopharmacology 2016, 41, 2352–2365. [Google Scholar] [CrossRef] [Green Version]
  303. Johnson, M.W.; Griffiths, R.R. Potential Therapeutic Effects of Psilocybin. Neurotherapeutics 2017, 14, 734–740. [Google Scholar] [CrossRef]
  304. Anderson, C.A.M.; Cobb, L.K.; Miller, E.R., III; Woodward, M.; Hottenstein, A.; Chang, A.R.; Mongraw-Chaffin, M.; White, K.; Charleston, J.; Tanaka, T.; et al. Effects of a Behavioral Intervention That Emphasizes Spices and Herbs on Adherence to Recommended Sodium Intake: Results of the SPICE Randomized Clinical Trial. Am. J. Clin. Nutr. 2015, 102, 671–679. [Google Scholar] [CrossRef] [Green Version]
  305. Avena, N.M.; Rada, P.; Hoebel, B.G. Evidence for Sugar Addiction: Behavioral and Neurochemical Effects of Intermittent, Excessive Sugar Intake. Neurosci. Biobehav. Rev. 2008, 32, 20–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  306. Bowen, S.; Cruz, S.L. Inhalants. In The Effects of Drug Abuse on the Human Nervous System; Madras, B., Kuhar, M., Eds.; Elsevier: Amsterdam, The Netherlands, 2014; pp. 553–574. [Google Scholar]
  307. Cohen, K.; Weinstein, A.M. Synthetic and Non-Synthetic Cannabinoid Drugs and Their Adverse Effects-A Review from Public Health Prospective. Front. Public Health 2018, 6, 162. [Google Scholar] [CrossRef] [PubMed]
  308. D’amanda, C.; Plumb, M.M.; Taintor, Z. Heroin Addicts with a History of Glue Sniffing: A Deviant Group within a Deviant Group. Int. J. Addict. 1977, 12, 255–270. [Google Scholar] [CrossRef] [PubMed]
  309. Elster, J. Strong Feelings: Emotion, Addiction, and Human Behavior; MIT Press: Cambridge, MA, USA, 2009. [Google Scholar]
  310. Fernández-Montalvo, J.; López-Goñi, J.J.; Arteaga, A. Violent Behaviors in Drug Addiction: Differential Profiles of Drug-Addicted Patients with and without Violence Problems: Differential Profiles of Drug-Addicted Patients with and without Violence Problems. J. Interpers. Violence 2012, 27, 142–157. [Google Scholar] [CrossRef] [Green Version]
  311. Filgueiras, A.R.; Pires de Almeida, V.B.; Koch Nogueira, P.C.; Alvares Domene, S.M.; Eduardo da Silva, C.; Sesso, R.; Sawaya, A.L. Exploring the Consumption of Ultra-Processed Foods and Its Association with Food Addiction in Overweight Children. Appetite 2019, 135, 137–145. [Google Scholar] [CrossRef]
  312. Freedman, A.M.; Fink, M.; Sharoff, R.; Zaks, A. Clinical Studies of Cyclazocine in the Treatment of Narcotic Addiction. Am. J. Psychiatry 1968, 124, 1499–1504. [Google Scholar] [CrossRef]
  313. German, C.L.; Fleckenstein, A.E.; Hanson, G.R. Bath Salts and Synthetic Cathinones: An Emerging Designer Drug Phenomenon. Life Sci. 2014, 97, 2–8. [Google Scholar] [CrossRef] [Green Version]
  314. Greenblatt, D.J.; Shader, R.I. Dependence, Tolerance, and Addiction to Benzodiazepines: Clinical and Pharmacokinetic Considerations. Drug Metab. Rev. 1978, 8, 13–28. [Google Scholar] [CrossRef]
  315. Isbell, H. Addiction to Barbiturates and the Barbiturate Abstinence Syndrome. Ann. Intern. Med. 1950, 33, 108–121. [Google Scholar]
  316. Katselou, M.; Papoutsis, I.; Nikolaou, P.; Spiliopoulou, C.; Athanaselis, S. A “Krokodil” Emerges from the Murky Waters of Addiction. Abuse Trends of an Old Drug. Life Sci. 2014, 102, 81–87. [Google Scholar] [CrossRef]
  317. Krebs, T.S.; Johansen, P.-Ø. Psychedelics and Mental Health: A Population Study. PLoS ONE 2013, 8, e63972. [Google Scholar] [CrossRef] [PubMed]
  318. Lange, W.R.; Fudala, P.J.; Dax, E.M.; Johnson, R.E. Safety and Side-Effects of Buprenorphine in the Clinical Management of Heroin Addiction. Drug Alcohol Depend. 1990, 26, 19–28. [Google Scholar] [CrossRef]
  319. Lopez-Lopez, D.E.; Saavedra-Roman, I.K.; Calizaya-Milla, Y.E.; Saintila, J. Food Addiction, Saturated Fat Intake, and Body Mass Index in Peruvian Adults: A Cross-Sectional Survey. J. Nutr. Metab. 2021, 2021, 9964143. [Google Scholar] [CrossRef] [PubMed]
  320. Morgan, W.W. Abuse Liability of Barbiturates and Other Sedative-Hypnotics. Adv. Alcohol Subst. Abuse 1990, 9, 67–82. [Google Scholar] [CrossRef]
  321. Pagonis, T.A.; Angelopoulos, N.V.; Koukoulis, G.N.; Hadjichristodoulou, C.S. Psychiatric Side Effects Induced by Supraphysiological Doses of Combinations of Anabolic Steroids Correlate to the Severity of Abuse. Eur. Psychiatry 2006, 21, 551–562. [Google Scholar] [CrossRef]
  322. Phillips, B.; Hajela, R.; Hilton, D.L., Jr. Sex Addiction as a Disease: Evidence for Assessment, Diagnosis, and Response to Critics. Sex. Addict. Compulsivity 2015, 22, 167–192. [Google Scholar] [CrossRef]
  323. Polosa, R.; Benowitz, N.L. Treatment of Nicotine Addiction: Present Therapeutic Options and Pipeline Developments. Trends Pharmacol. Sci. 2011, 32, 281–289. [Google Scholar] [CrossRef] [Green Version]
  324. Real, T.; Cruz, S.L.; Medina-Mora, M.E.; Robles, R.; González, H. Inhalant Addiction. In Textbook of Addiction Treatment; Springer International Publishing: Cham, Switzerland, 2021; pp. 281–306. [Google Scholar]
  325. Satel, S. Is Caffeine Addictive?—A Review of the Literature. Am. J. Drug Alcohol Abuse 2006, 32, 493–502. [Google Scholar] [CrossRef]
  326. Schifano, F. Potential Human Neurotoxicity of MDMA (‘Ecstasy’): Subjective Self-Reports, Evidence from an Italian Drug Addiction Centre and Clinical Case Studies. Neuropsychobiology 2000, 42, 25–33. [Google Scholar] [CrossRef]
  327. Soto-Escageda, J.A.; Estañol Vidal, B.; Vidal-Victoria, C.A.; Michel Chávez, A.; Sierra-Beltran, M.A.; Bourges-Rodríguez, H. Does Salt Addiction Exist? Salud Ment. 2016, 39, 175. [Google Scholar] [CrossRef] [Green Version]
  328. Spring, B.; Schneider, K.; Smith, M.; Kendzor, D.; Appelhans, B.; Hedeker, D.; Pagoto, S. Abuse Potential of Carbohydrates for Overweight Carbohydrate Cravers. Psychopharmacology 2008, 197, 637–647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  329. Terenius, L. Alcohol Addiction (Alcoholism) and the Opioid System. Alcohol 1996, 13, 31–34. [Google Scholar] [CrossRef]
  330. Braverman, E.R.; Blum, K.; Loeffke, B.; Baker, R.; Kreuk, F.; Yang, S.P.; Hurley, J.R. Managing Terrorism or Accidental Nuclear Errors, Preparing for Iodine-131 Emergencies: A Comprehensive Review. Int. J. Environ. Res. Public Health 2014, 11, 4158–4200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  331. Sethi, R.; Hoang, N.; Ravishankar, D.A.; McCracken, M.; Manzardo, A.M. Kratom (Mitragyna Speciosa): Friend or Foe? Prim. Care Companion CNS Disord. 2020, 22, 27410. [Google Scholar] [CrossRef]
  332. Jowitt, D.M.A. Kava in the Pacific Islands: A Contemporary Drug of Abuse? Drug Alcohol Rev. 2000, 19, 217–227. [Google Scholar]
  333. Nawaz, H.; Chan, W.; Abdulrahman, M.; Larson, D.; Katz, D.L. Self-Reported Weight and Height: Implications for Obesity Research. Am. J. Prev. Med. 2001, 20, 294–298. [Google Scholar] [CrossRef]
  334. Beck, O.; Stephanson, N.; Sandqvist, S.; Franck, J. Detection of Drugs of Abuse in Exhaled Breath Using a Device for Rapid Collection: Comparison with Plasma, Urine and Self-Reporting in 47 Drug Users. J. Breath Res. 2013, 7, 026006. [Google Scholar] [CrossRef] [Green Version]
  335. Gilligan, C.; Anderson, K.G.; Ladd, B.O.; Yong, Y.M.; David, M. Inaccuracies in Survey Reporting of Alcohol Consumption. BMC Public Health 2019, 19, 1639. [Google Scholar] [CrossRef] [Green Version]
  336. Large, M.M.; Smith, G.; Sara, G.; Paton, M.B.; Kedzior, K.K.; Nielssen, O.B. Meta-Analysis of Self-Reported Substance Use Compared with Laboratory Substance Assay in General Adult Mental Health Settings: Self-Reported Substance Use Compared with Laboratory Substance Assay. Int. J. Methods Psychiatr. Res. 2012, 21, 134–148. [Google Scholar] [CrossRef]
  337. Harris, K.M.; Griffin, B.A.; McCaffrey, D.F.; Morral, A.R. Inconsistencies in Self-Reported Drug Use by Adolescents in Substance Abuse Treatment: Implications for Outcome and Performance Measurements. J. Subst. Abuse Treat. 2008, 34, 347–355. [Google Scholar] [CrossRef] [Green Version]
  338. Comings, D.E.; MacMurray, J.; Johnson, P.; Dietz, G.; Muhleman, D. Dopamine D2 Receptor Gene (DRD2) Haplotypes and the Defense Style Questionnaire in Substance Abuse, Tourette Syndrome, and Controls. Biol. Psychiatry 1995, 37, 798–805. [Google Scholar] [CrossRef]
  339. Kandel, D.; Kandel, E. The Gateway Hypothesis of Substance Abuse: Developmental, Biological and Societal Perspectives. Acta Paediatr. 2015, 104, 130–137. [Google Scholar] [CrossRef] [PubMed]
  340. Wade, N.E.; Bagot, K.S.; Tapert, S.F.; Gruber, S.A.; Filbey, F.M.; Lisdahl, K.M. Cognitive Functioning Related to Binge Alcohol and Cannabis Co-Use in Abstinent Adolescents and Young Adults. J. Stud. Alcohol Drugs 2020, 81, 479–483. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Clinical electroencephalography [257]. (A): Control/baseline. (B): Right frontal-temporal abnormalities typical of individuals with mood swings, cognitive decline, anxiety, and depression without SUD. (C): Significant SUD with worsening bitemporal damage [247,257,266,288,289,290,291]. (with permission from publisher).
Figure 1. Clinical electroencephalography [257]. (A): Control/baseline. (B): Right frontal-temporal abnormalities typical of individuals with mood swings, cognitive decline, anxiety, and depression without SUD. (C): Significant SUD with worsening bitemporal damage [247,257,266,288,289,290,291]. (with permission from publisher).
Ijerph 19 05480 g001
Table 1. Common medical comorbidities of SUD. 1
Table 1. Common medical comorbidities of SUD. 1
NeurologicDementia, Wernicke–Korsakoff Syndrome, Seizures, Cerebrovascular Accident, Cerebrovascular Disease, Polyneuropathy, Encephalopathy, Hepatic Encephalopathy, Head trauma, Sleep Disorders, Multiple Sclerosis, Neurodegenerative disorders, Neonatal Abstinence Syndrome
PsychiatricAnxiety, Depression, Bipolar, Post Traumatic Stress Disorder, Attention Deficit Hyperactivity Disorder, Attention Deficit Disorder, Psychosis, Personality Disorders, Chronic Pain, Suicide, Sleep Disorder (decreased duration, REM, and CSF brain wash)
CardiovascularHypertension, Coronary Atherosclerosis, Arrhythmias, Cardiomyopathy, Ischemic Heart Disease, Congestive Heart Failure, Myocardial infarction, Peripheral Vascular Disease
PulmonaryPneumonia, Aspiration Pneumonia, Asthma, Allergic Rhinitis, Toxic Rhinitis, Chronic Obstructive Pulmonary Disease, Lung Cancer
GastrointestinalEsophagitis, Mallory–Weiss Syndrome, Boerhaave Syndrome, Gastritis, Peptic Ulcer, Gallbladder Disease, Pancreatitis, Cirrhosis, Alcoholic Liver Disease, Nonalcoholic Fatty Liver Disease, Mild/moderate, Severe Liver Disease, Portal Hypertension, Intestinal Ischemia, Gastrointestinal Perforation, Inflammatory Bowel Disease
EndocrineDiabetes, Obesity, Metabolic Syndrome, Hypothyroidism, Neuroendocrine abnormalities
InfectiousHepatitis, Endocarditis, Bacterial Pneumonia, Tuberculosis, Skin Infections, Sexually Transmitted Diseases, Human Immunodeficiency Virus, Acquired Immunodeficiency Syndrome, Arthritis
HematologicAnemia, White Blood Cell Disorders, Platelet Disorders, Splenomegaly, Hyposplenism, Coagulopathy
NephrologyRenal disease, Renal Failure, Fluid and Electrolyte Disorders
GenitourinaryUrinary Retention, Erectile Dysfunction
MusculoskeletalOsteoporosis/penia, Sarcoporosis/penia, Fragility, Fibromyalgia, Temporomandibular Joint and Muscle disorders, Paralysis
DermatologicScabies, Jaundice, Pruritus, Urticaria, Hyperpigmentation, Spider Telangiectasias, Angiomas, Caput Medusas. Flushing, Palmar Erythema, Psoriasis, Porphyria Cutanea Tarda, Leukonychia, Rhytids, Drug Injection Lesions (track marks)
SocialTrauma, Violent Behaviors, Criminal Behavior, Prison, Divorce, Homelessness, Internet Gaming
OtherScleral Icterus, Cancer, Weight Loss, Inflammation
Ref: [51,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167]
1 Charlson–Deyo and Elixhauser-van Walraven (has SUD as a factor) Comorbidity Index scores consolidate the common medical comorbidities table (Table 1) into high-risk diagnoses, but if used alone miss 90% of secondary comorbidities caused by SUD [36,51]. REM: Rapid Eye Movement; CSF: Cerebral Spinal Fluid.
Table 2. SUD premorbid states.
Table 2. SUD premorbid states.
CognitiveADHD, ADD, Impaired Memory, Impaired Judgment
Neuropsychiatric IllnessAny Psychiatric Illness, Head Concussion, Trauma, Chronic Traumatic Encephalopathy (CTE), Birth Injury, Sleep Disorders
SocialLow Education Attainment, Disturbed Family Life, Family History of Addiction, Abnormal Genetic Addiction Risk Scores, Amphetamine Use, Poverty, School Truancy, Unintended Pregnancy, Culture
REF: [32,33,34,53,169,170,171,172,173,174,175,176,177,178,179,180]
Table 3. Parallel pattern to manage brain and cardiac disease. 1
Table 3. Parallel pattern to manage brain and cardiac disease. 1
Core Brain Domains/TestsCore Cardiac Domains/Tests
  • Memory: CNSVS
  • Attention: TOVA
  • Neuropsychiatric: MCMI-III
  • Genetic: GARS
  • Imaging: Electrophysiology, qEEG/p300/EP
  • Blood pressure
  • Blood work: cholesterol, CRP, etc.
  • Electrophysiology: EKG
  • Echocardiogram: Valves, Ejection fraction
  • Imaging: CT angiogram
Supplemental Brain TestingSupplemental Cardiac Testing
  • Memory: WMS, MCI Screening, MMSE
  • Attention: Connors, ADD Checklist
  • Neuropsychiatric: TT, MBTI, MMPI
  • Imaging: MRI, PET, SPECT
  • Electrophysiological: Holter, EP testing
  • Echocardiogram: Transesophageal
  • Blood work: BNP, cardiac enzymes, etc.
  • Imaging: Catheterization, MRI, PET
1 CNSVS: Central Nervous System Vital Signs, TOVA: Test of Variables of Attention, MCMI-III: Millon Clinical Multiaxial Inventory III, GARS: Genetic Addiction Risk Score, qEEG: Quantitative Electroencephalogram, EP: Evoked Potential, WMS: Wechsler Memory Scale, MCI: Mild Cognitive Impairment, MMSE: Mini-Mental State Examination, ADD: Attention Deficit Disorder, TT: Type and Temperament, MBTI: Myers–Briggs Type Indicator, MMPI: Minnesota Multiphasic Personality Inventory, MRI: Magnetic Resonance Imaging, PET: Positron Emission Tomography, SPECT: Single-Photon Emission Computed Tomography, CRP: C-Reactive Protein, BNP: B-type Natriuretic Peptide.
Table 4. Common areas of brain atrophy in SUD and MCI.
Table 4. Common areas of brain atrophy in SUD and MCI.
AmygdalaMidbrain
Anterior cingulate cortexNucleus Accumbens
Basal GangliaOccipital cortex
CerebellumOccipitoparietal cortex
Cingulate gyrusOrbitofrontal cortex
Extended AmygdalaParahippocampal gyrus
Frontal cingulateParietal cortex
Frontal CortexPrefrontal Cortex
Globus pallidusPulvinar
InsulaPutamen
Left temporal gyrusSuperior frontal gyrus
Medial Frontal CortexThalamus
MesencephalonVentral Tegmental Area
REF: [197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226]
Table 5. Neuropsychiatric consequences of SUD. 1
Table 5. Neuropsychiatric consequences of SUD. 1
MemoryAttentionNeuropsychiatricIQ/Cognitive Considerations
↓Composite Memory
↓Verbal Memory
↓Visual Memory
↓Visual Immediate
↓Visual Delay
↓Auditory Immediate
↓Auditory Delay
↓Auditory Recognition Delay
↓General Memory
↓Working Memory
↓Spatial Memory
↓Declarative Memory
↓Visuoperception
↓Sensory Memory
↓Episodic Memory
↓Visuo-constructional abilities
↓Prospective Memory
↓Retro Memory
↓Attention
↓Complex Attention
↓Simple Visual Attention
↓Reaction Time
↓Response Time
↓Psychomotor Speed
↓Processing Speed
↓Motor Speed
↓Executive Function
↓Delayed Recall
↓Inattentiveness
↓Manual Dexterity
↓Inhibitory Control
↓Temporal Processing
↓Cognitive Flexibility

↑Impulsivity
↑Omission Errors
↑Commissions Errors

Response Time Variability
Schizoid
Avoidant
Depressive
Dependent
Histrionic
Narcissistic
Antisocial
Sadistic
Compulsive
Negativistic
Masochistic
Schizotypal
Borderline
Paranoid
Anxiety
Somatoform
Bipolar Manic
Dysthymia
Alcohol Dependence
Post-Traumatic Stress
Thought Disorder
Major Depression
Delusional Disorder
↓IQ
↓Verbal IQ
↓Performance IQ
↓Abstract IQ
↓General Cognitive Functioning
↓General Intelligence
↓Reasoning
REF: [34,198,246,251,252]
1 The range of neuropsychiatric consequences of SUD are extremely diverse and without uniformity of testing cannot be measured. The downward-facing arrows indicate decreases, and the upward-facing arrows indicate increases.
Table 6. Sample MCMI-III score report. 1
Table 6. Sample MCMI-III score report. 1
CategoryBR ScoreDiagnostic Scales
Modifying Indices93Disclosure
20Desirability
90Debasement
Clinical Personality Patterns62Schizoid
83Avoidant
83Depressive
93Dependent
12Histrionic
40Narcissistic
66Antisocial
57Sadistic
16Compulsive
94Negativistic
65Masochistic
Severe Personality Pathology65Schizotypal
93Borderline
68Paranoid
Clinical Syndromes97Anxiety
66Somatoform
71Bipolar: Manic
88Dysthymia
68Alcohol Dependence
76Drug Dependence
76Post-Traumatic Stress
Severe Clinical Syndromes70Thought disorder
100Major Depression
63Delusional Disorder
1 Significant scores are BR > 75 [256].
Table 7. Brain neuroimaging. 1
Table 7. Brain neuroimaging. 1
Brain Imaging StudiesAlterations in Brain Activation Patterns While Performing Cognitive Tasks
EEG/qEEG
p300/EP
(electrophysiology)
  • Delayed p300 latency
  • Decreased voltage of p300
  • Abnormalities in auditory and visual evoked potentials
  • Increased theta waves, decreased alpha and beta waves
  • Abnormal polysomnography, i.e., increased nocturnal movement, decreased REM, decrease sleep efficiency
SPECT
  • Blood flow single-photon emission computed tomography
  • Decreased prefrontal lobe and temporal lobe circulation
  • Decreased cerebral circulation
fMRI
  • Abnormal diffuser tensor imaging
  • Abnormal fiber connections
  • Abnormal neuropsychological tasks
  • Hypo-activation of neuronal networks
  • Prefrontal, frontal, parietal regions
MRI
  • Smaller brain volume in 5 subcortical areas including amygdala, hippocampus, etc.
  • SUD may have increased cortical thickness
  • Brain Atrophy multiple regions (see Table 4)
  • White Matter Microstructure
PET
  • Abnormal metabolism of dopamine and its transporters
  • Abnormal binding to D2 receptors meg phase increases coherence beta gamma
-
Abnormalities in working memory
-
Anomalies in auditory and visual processing
REF: [198,251,257,264,265,266,267,268,269,270,271,272,273,274,275,276,277]
1 Brain Health MAP imaging is very diverse, but over the past 40 years, electrophysiology has become the standard and the most cost-effective. Evoked potential = EP.
Table 8. The gateway of potential addictive substances and behaviors. 1
Table 8. The gateway of potential addictive substances and behaviors. 1
Illegal SubstancesLegal SubstancesLegal FoodsAbnormal Behavior
Synthetic Cannabinoid
(Marijuana, CBD, Spice)
Synthetic Cannabinoid
(Marijuana, CBD)
CaffeineGambling
InhalantsNicotineSugars Internet
Anabolic SteroidsAlcoholCarbohydratesSex
KetamineAnabolic SteroidsSaltShopping
CocainePrescribed OpioidsFats, Trans, TallowThoughts/Feelings
HeroinBenzodiazepineCharcoaled foodViolence
Amphetamines (MDMA, Ecstasy)KetamineSpiced foods
Narcotics (Crocodil)NarcoticsCanned Foods
Psychedelics (LSD, Salvia, Mushrooms)BarbituratesPackaged Foods
Synthetic Cathinones (Bath Salts, Flakka)Sedative/hypnotics,Processed Foods
Miscellaneous (Kratom, Quaaludes, New Market Designer Drugs)Miscellaneous (Kava, Kratom, Glue, Gasoline, etc.)
REF: [2,13,173,304,305,306,307,308,309,310,311,312,313,314,315,316,317,318,319,320,321,322,323,324,325,326,327,328,329,330,331,332]
1 Every drug has multiple methods of entry, e.g., vaping, snorting, combustible, and liquid, etc.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Braverman, E.R.; Dennen, C.A.; Gold, M.S.; Bowirrat, A.; Gupta, A.; Baron, D.; Roy, A.K.; Smith, D.E.; Cadet, J.L.; Blum, K. Proposing a “Brain Health Checkup (BHC)” as a Global Potential “Standard of Care” to Overcome Reward Dysregulation in Primary Care Medicine: Coupling Genetic Risk Testing and Induction of “Dopamine Homeostasis”. Int. J. Environ. Res. Public Health 2022, 19, 5480. https://doi.org/10.3390/ijerph19095480

AMA Style

Braverman ER, Dennen CA, Gold MS, Bowirrat A, Gupta A, Baron D, Roy AK, Smith DE, Cadet JL, Blum K. Proposing a “Brain Health Checkup (BHC)” as a Global Potential “Standard of Care” to Overcome Reward Dysregulation in Primary Care Medicine: Coupling Genetic Risk Testing and Induction of “Dopamine Homeostasis”. International Journal of Environmental Research and Public Health. 2022; 19(9):5480. https://doi.org/10.3390/ijerph19095480

Chicago/Turabian Style

Braverman, Eric R., Catherine A. Dennen, Mark S. Gold, Abdalla Bowirrat, Ashim Gupta, David Baron, A. Kenison Roy, David E. Smith, Jean Lud Cadet, and Kenneth Blum. 2022. "Proposing a “Brain Health Checkup (BHC)” as a Global Potential “Standard of Care” to Overcome Reward Dysregulation in Primary Care Medicine: Coupling Genetic Risk Testing and Induction of “Dopamine Homeostasis”" International Journal of Environmental Research and Public Health 19, no. 9: 5480. https://doi.org/10.3390/ijerph19095480

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