Ds 260 Sample: Fill & Download for Free

GET FORM

Download the form

How to Edit The Ds 260 Sample easily Online

Start on editing, signing and sharing your Ds 260 Sample online refering to these easy steps:

  • Push the Get Form or Get Form Now button on the current page to jump to the PDF editor.
  • Wait for a moment before the Ds 260 Sample is loaded
  • Use the tools in the top toolbar to edit the file, and the change will be saved automatically
  • Download your completed file.
Get Form

Download the form

The best-rated Tool to Edit and Sign the Ds 260 Sample

Start editing a Ds 260 Sample right now

Get Form

Download the form

A quick direction on editing Ds 260 Sample Online

It has become very easy just recently to edit your PDF files online, and CocoDoc is the best tool you have ever used to have some editing to your file and save it. Follow our simple tutorial to start!

  • Click the Get Form or Get Form Now button on the current page to start modifying your PDF
  • Add, change or delete your content using the editing tools on the toolbar on the top.
  • Affter altering your content, add the date and make a signature to complete it perfectly.
  • Go over it agian your form before you click on the button to download it

How to add a signature on your Ds 260 Sample

Though most people are adapted to signing paper documents with a pen, electronic signatures are becoming more common, follow these steps to sign PDF online for free!

  • Click the Get Form or Get Form Now button to begin editing on Ds 260 Sample in CocoDoc PDF editor.
  • Click on the Sign tool in the tools pane on the top
  • A window will pop up, click Add new signature button and you'll have three ways—Type, Draw, and Upload. Once you're done, click the Save button.
  • Drag, resize and settle the signature inside your PDF file

How to add a textbox on your Ds 260 Sample

If you have the need to add a text box on your PDF for customizing your special content, do some easy steps to carry it out.

  • Open the PDF file in CocoDoc PDF editor.
  • Click Text Box on the top toolbar and move your mouse to position it wherever you want to put it.
  • Write in the text you need to insert. After you’ve input the text, you can use the text editing tools to resize, color or bold the text.
  • When you're done, click OK to save it. If you’re not happy with the text, click on the trash can icon to delete it and do over again.

A quick guide to Edit Your Ds 260 Sample on G Suite

If you are looking about for a solution for PDF editing on G suite, CocoDoc PDF editor is a commendable tool that can be used directly from Google Drive to create or edit files.

  • Find CocoDoc PDF editor and establish the add-on for google drive.
  • Right-click on a PDF document in your Google Drive and click Open With.
  • Select CocoDoc PDF on the popup list to open your file with and allow access to your google account for CocoDoc.
  • Modify PDF documents, adding text, images, editing existing text, mark up in highlight, fullly polish the texts in CocoDoc PDF editor before saving and downloading it.

PDF Editor FAQ

What percentage of depression has been solidly linked to genetic mutations (not including polymorphisms)?

What percentage of depression has been solidly linked to genetic mutations (not including polymorphisms)?The cause of depression, and other emotional maladies, is a complex interplay between nature and nurture. Genetics, upbringing, culture, diet, stress levels, and epigenetics all play a role.I don't think you can put a firm number on it.Anyone can get depressed.The same might be said of “Resiliance”, our ability to resist having our internal homeostasis disturbed.Some people just seem better able to deal with the ups and downs.Certainly there are things we can do, like exercise, healthy diet, emotional counseling, medications, etc. that can help.Twin studies suggest 40% or so genetic linkage. But, this is highly contested.However, as with most attempts to find a direct genetic link to disease, there are too many variables.It's too complex for simple answers, or simple solutions.My personal take on the subject, having had my own problems with depression, is that depression is a sign of internal toxicity.Internal toxicity can come from many sources.Toxins in food, toxic drugs, toxic environment, toxic relationships, toxic work, toxic attitudes, toxic people, toxic self loathing... Some things poison your body, your mind, your spirit.The best things you can do for yourself is to remove or reduce exposure to toxic things.Figure out what is poisoning you, and get it out of your life as best you canDepression is a symptom, not a disease.Which is why I think there is no answer to this question.Here is a good starter article cribbed from pubmed:Overview of the Genetics of Major Depressive DisorderOverview of the Genetics of Major Depressive DisorderFalk W. LohoffAbstractMajor depressive disorder (MDD) is a common psychiatric illness with high levels of morbidity and mortality. Despite intensive research during the past several decades, the neurobiological basis and pathophysiology of depressive disorders remain unknown. Genetic factors play important roles in the development of MDD, as indicated by family, twin, and adoption studies, and may reveal important information about disease mechanisms. This article describes recent developments in the field of psychiatric genetics, with a focus on MDD. Early twin studies, linkage studies, and association studies are discussed. Recent findings from genome-wide association studies are reviewed and future directions discussed. Despite all efforts, thus far, no single genetic variation has been identified to increase the risk of depression substantially. Genetic variants are expected to have only small effects on overall disease risk, and multiple genetic factors in conjunction with environmental factors are likely necessary for the development of MDD. Future large-scale studies are needed to dissect this complex phenotype and to identify pathways involved in the etiology of MDD.Keywords: Depression, Mood disorder, Linkage, Association, Genetics, SNPIntroductionMajor depressive disorder (MDD) is a common psychiatric illness with high levels of morbidity and mortality. It is estimated that 10% to 15% of the general population will experience clinical depression during their lifetime [1], and 5% of men and 9% of women will experience a depressive disorder in a given year, according to the World Health Organization [2]. Genetic factors play important roles in the development of MDD, as indicated by family, twin, and adoption studies. Twin studies suggest a heritability of 40% to 50%, and family studies indicate a twofold to threefold increase in lifetime risk of developing MDD among first-degree relatives. This degree of familial aggregation, coupled with the high heritability from twin studies, generated optimism that molecular genetic techniques would reveal genes of substantial influence on MDD risk. Unfortunately, gene localization and identification has been a slow, labor-intensive process. Genetic investigators have encountered similar frustrations with other common complex traits (eg, asthma, hypertension, and diabetes mellitus).The major impediments to mood disorder gene localization and identification are as follows: 1) no single gene is necessary and sufficient for MDD; 2) each susceptibility gene contributes a small fraction of the total genetic risk; and 3) complex genetic heterogeneity, meaning that multiple partially overlapping sets of susceptibility genes (which interact with the environment) can predispose individuals to similar syndromes that are indistinguishable on clinical grounds. This article provides an overview of the current efforts to identify genetic risk factors for MDD.Twin and Family StudiesEvidence for a genetic component to mood disorders has been documented consistently using family, twin, and adoption studies. The first genetic studies of mood disorders were conducted more than 70 years ago and included assessment of concordance rates for monozygotic and dizygotic twins with mood disorders [3]. These early studies did not distinguish between bipolar depression and MDD-recurrent unipolar (MDD-RU). A recent review of twin studies in MDD-RU estimated heritability at 37%, with a substantial component of unique individual environmental risk but little shared environmental risk [4].Family studies of MDD-RU have shown that first-degree relatives of MDD-RU probands are at increased risk of MDD-RU disorders compared with first-degree relatives of control probands [5]. There was a twofold to fourfold increased risk of MDD-RU among the first-degree relatives of MDD-RU probands. Characteristics of MDD-RU disorders that yield a more heritable phenotype include early onset (ie, before age 30 years) and a high degree of recurrence. A third characteristic that may identify a separate group of disorders is the presence of psychosis. Additional genetic subtypes of MDD-RU may be identified through examination of comorbidities with panic disorder, other anxiety disorders, and alcoholism.Linkage StudiesBecause of the epidemiologic evidence for a genetic component of MDD, the field had hoped that the identification of genetic risk factors would be straightforward. The first wave of comprehensive investigations of the genetic origins of MDD used linkage studies. Linkage studies have proven to be successful in identifying genetic risk factors for rare mendelian disorders with high penetrance, such as autosomal-dominant inherited forms of epilepsy or cystic fibrosis. The term linkage refers to the observation that two genetic loci found near each other on the same chromosome tend to be inherited together more often than expected by chance within families. Two such loci are said to be linked. The key concept of linkage is that chromosomal fragments that might harbor vulnerability genes are inherited with an illness more often then expected by chance in families.Although some linkage studies in MDD have suggested several regions in the genome that might harbor risk alleles, findings have been inconsistent, and thus far, no established universal genetic risk factor or causative gene for depression has been identified. Although these initial results from linkage studies in MDD seem rather disappointing, they emphasize that MDD is a complex disorder with a complex mode of inheritance, that multiple genes with small effects likely are involved, and that identifying genetic factors is complicated by a significant gene–environment component.Several MDD linkage scans have been conducted during the past several years and are reviewed in detail elsewhere [6]; however, only a few studies had a sufficient number of affected individuals (>100). Holmans et al. [7] reported on the first phase of a multisite collaborative effort (Recurrent Early-Onset Depression [GenRED] sample). The sample consisted of 297 informative multiplex families (containing 685 informative affected relative pairs, 555 sibling pairs, and 130 other pair types). Affected cases had MDD-RU with onset before 31 years of age for probands or 41 for other affected relatives. The mean age at onset was 18.5 years, and the mean number of depressive episodes was 7.3, indicating a highly recurrent form of illness. Families were excluded if there was a first-degree or second-degree relative with bipolar disorder (BPD) [7]. Linkage was observed on chromosome 15q25.3-26.2 (empiric genome-wide P=0.023). The linkage was not sex specific. This was the sole significant linkage peak observed by this group. In the complete sample of 656 families, genome-wide suggestive linkage was confirmed on chromosome 15q and also observed on chromosomes 17p and 8p in a planned second analysis accounting for the sex of each pair of relatives [8]. Fine mapping of the 15q region demonstrated further evidence of linkage [9].Abkevich et al. [10] reported a genome scan on 110 Utah pedigrees (each with at least four affected individuals), in which there were 784 individuals with MDD-RU, 161 with single-episode MDD, and 162 with BPD who were also considered affected. They observed a highly significant linkage signal at 12q23 [10], confirming a previously identified BPD locus. No other linkage peaks approached statistical significance. It is probable that this study detected the same BPD 12q23 locus, even though the families were ascertained from an MDD-RU proband, because most kindreds probably did have at least one BPD individual. These results confirm the findings of family and twin studies suggesting genetic overlap between BPD and MDD-RU disorders, and this study identifies the 12q23 region as a locus that increases risk of both BPD and MDD-RU disorders.Camp et al. [11] reanalyzed the large Utah pedigrees and excluded relatives with BPD. They considered three alternative phenotypes (MDD age at onset earlier than 31 years of age, MDD or anxiety, MDD and anxiety) and identified regions with at least suggestive genome-wide evidence for linkage on chromosomes 3centr, 7p, and 18q [11]. Interestingly, the region identified on 18q with MDD and anxiety is also a well-replicated linkage finding in BPD.Another recent genome-wide linkage scan was carried out using 497 sibling pairs concordant for recurrent depression, excluding BPD. The advantage of affected sibling pair design is that it does not require knowledge of mode of inheritance and increased power under certain conditions. Suggestive evidence of linkage was observed on chromosomes 1p36, 12q23.3-q24.11, and 13q31.1-q31.3 [12]. The 12q locus was previously implicated in linkage studies of unipolar [10] and bipolar disorders, while the 13q peak lies within a region previously linked strongly to panic disorder [13]. Middeldorp et al. [14] used 110 Australian and 23 Dutch MDD pedigrees with at least two affected siblings. The scan identified a region on chromosome 17 that includes the gene encoding the serotonin transporter (SLC6A4). Follow-up genotyping failed to identify the previously implicated promoter length polymorphism in SLC6A4 to be associated with MDD, suggesting that another SLC6A4 variant or a polymorphism in a different gene might contribute to this signal. Interestingly, they reported chromosome 8 as their most promising finding because this region has also been implicated in two previous linkage studies of personality traits, including harm avoidance [15] and neuroticism [16]. Although linkage efforts in MDD have not identified universal risk genes yet, they have provided insights into the genomic regions that might harbor genetic susceptibility factors.Candidate Gene StudiesCandidate gene studies of unipolar depression traditionally have received less attention in the past compared with those of BPD and schizophrenia. Likely reasons for this discrepancy might be practical limitations given the much smaller expected effect size and a more heterogeneous clinical phenotype. However, with increasing sample sizes, the literature is developing rapidly. As with other complex psychiatric disorders, there is no universal susceptibility gene for MDD. It can be expected that multiple genes with small effect sizes contribute to depression. Several candidate genes show promising preliminary results and are worth mentioning. Most candidate genes are studied using a case-control association study design. The basic principle of genetic association studies is that a genetic variant(s) is investigated in a group of cases and controls. By determining the allele or genotype frequencies and comparing them statistically, the probability that a genetic polymorphism is more frequent in one group than the other can be investigated. Hypotheses are generated based on the concept that specific variants increase or decrease risk of a certain phenotype. Genetic variants for study are usually selected based on an a priorihypothesis, such as neurobiologic plausibility (eg, serotonin transporter for antidepressants) or genomic location of a candidate gene (eg, in a linkage peak). More recently, “hypothesis-free” designs have been promoted with the advance of genome-wide association studies (GWAS), which space genetic markers across the whole genome based on linkage disequilibrium patterns and are discussed later; however, the resources required to conduct GWAS, including technological and clinical resources, are considerable and remain prohibitive in many instances. It is important to note that candidate gene association studies have several limitations. Such limitations include clinical and diagnostic heterogeneity, low statistical power if sample sizes are small, often-limited biological evidence of candidate gene selection, and unknown functional relevance of tested single nucleotide polymorphisms (SNPs), as well as population stratification within the sample leading to spurious positive findings or false-negative associations. Despite these obstacles, several candidate genes deserve mention, as they have been suggested repeatedly to be implicated in MDD.Serotonin Transporter (5HTT/SLC6A4) and Serotonin Receptor 2A (HTR2A)The serotonin transporter gene and genes involved in the serotonergic system are candidate genes for susceptibility to depression given that many antidepressant medications act on these systems. Several studies have implicated the serotonin transporter gene (SLC6A4) in MDD [17–19]. A 44-bp repeat polymorphism in the promoter region of the gene (5-HTTLPR) has been shown to influence expression levels of the serotonin transporter in vitro [20], thus making this functional variant a logical candidate for investigation in MDD. This polymorphism is the most studied genetic variant in psychiatric genetics to date. Similar to candidate gene studies in other complex psychiatric disorders, there have been some positive reports and some negative findings; additional studies are needed to dissect the exact role of this gene variant in the etiology of depressive disorders [21, 22•, 23].Because the serotonin transporter gene encodes a direct target for antidepressant medications, there has been great interest in correlating genetic variation with pharmacologic treatment response [24], and the field of pharmacogenetics is rapidly developing. Results have been similarly mixed, with some studies showing a statistically significant effect of the polymorphism and others failing to do so. A recent meta-analysis of 15 published studies concluded that there was a significant association between the L allele and better treatment response to selective serotonin reuptake inhibitors [25]. Interestingly, the association between the L allele and early antidepressant treatment response was the most robust finding in this meta-analysis, suggesting that the 5-HTTLPR might predict not only treatment response but also the time course of response and remission. A recent large-scale study using DNA samples from 1,953 patients with MDD who were treated with citalopram in the Sequenced Treatment Alternatives for Depression (STAR*D) trial investigated genetic predictors of treatment response [26]. The authors could not find evidence for 5HTT variation influencing treatment response; however, they reported a significant effect with a marker in the serotonin receptor gene HTR2A and treatment outcome. As expected for a single gene, the clinical impact of HTR2A on treatment outcome is modest. Although these studies face similar complexities and obstacles as disease candidate gene studies, this pharmacogenetic approach likely will yield robust results in the near future. Case-control association studies of the serotonin receptor gene HTR2A and major depression have yielded similar mixed results as for the serotonin transporter gene [23]. The European consortium project Genome-based Therapeutic Drugs for Depression (GENDEP) recently published its initial results. This is the first large-scale, multicenter human pharmacogenomics study focused on the prediction of therapeutic response to antidepressant drugs and adverse effects [27]. This open-label, flexible-dose, multicenter trial included 760 patients with MDD who were treated with citalopram or nortriptyline for 12 weeks. Initial analysis of ten candidate genes involved in serotonin, norepinephrine, neurotrophic, and glucocorticoid signaling revealed an association between treatment response to escitalopram and several variants in the serotonin receptor gene (HTR2A), with one marker (rs9316233) explaining 1.1% of the response variance. SNPs in the norepinephrine transporter gene (SLC6A2) predicted response to nortriptyline, and variants in the glucocorticoid receptor gene (NR3C1) predicted response to both antidepressants [28]. These data further support a role for the influence of genetic variants on treatment response to antidepressant drugs. Because single-marker analysis only explains a small fraction of the variance, future studies will have to use a multiple variant approach to find clinically meaningful genetic prediction algorithms.Gene–environment interaction studies have received increasing attention, particularly for MDD, given the robust correlation between stressful life events and risk of developing depressive symptoms [17, 29]. In a population-based study, Caspi and colleagues [21] noted that individuals with one or more copies of the short allele of the 5HTT promoter variants were at increased risk of depression depending on the occurrence of adverse life events. This article describes a plausible gene–environment interaction that may help explain the conflicting results for the 5HTT promoter variant noted above. Positive and negative replication studies have demonstrated the complexity of detecting these effects. Similar gene–environment interactions have been demonstrated for variants in the HTR2A gene and childhood maternal nurturance and depressive symptoms in adulthood [30]. Future genetic studies of depression will need to pay close attention to these gene–environment interactions.Brain-Derived Neurotrophic FactorGrowing evidence suggests an important role for brain-derived neurotrophic factor (BDNF) in affective disorder [31, 32]. Preclinical animal studies have consistently documented a role of BDNF in neurogenesis [33], and animal models of depression further substantiate a role of BDNF in mood disorders. Decreased BDNF levels in the hippocampus have been reported in animals exposed to chronic stress [34]. Interestingly, administration of anti-depressants increased hippocampal BDNF, preventing the stress-induced decrease [35]. These findings are intriguing given the hippocampal volume loss observed in mood disorders and decreased BDNF serum levels in individuals with mood disorders or depressive personality traits [32]. Based on these convergent preclinical and clinical data, the BDNF gene represents a logical target for genetic investigations of mood disorders.Although there is stronger literature support for a genetic association between the Val66Met polymorphism in the BDNF gene and BPD, several studies have also investigated this SNP in MDD. Results have been similarly mixed, as with most other candidate genes for depression. Schumacher et al. [36] examined 465 individuals with MDD but did not find a significant association with the Val66Met polymorphism. However, there was evidence of a haplotypic association [36]. Surtees and colleagues [37] failed to detect an association of the Val66Met polymorphism in 1,214 individuals with a history of MDD, while studies of this polymorphism in Asian populations yielded inconsistent results. Despite these mixed and negative results, interpretation of these data should be carried out with caution given the complex structure of the gene and the fact that most studies have only investigated the Val66Met SNP. It is likely that other variation in the BDNF gene influences susceptibility to depression [38].Tryptophan HydroxylaseTryptophan hydroxylase is the rate-limiting enzyme in brain serotonin synthesis. The discovery of a new brain-specific isoform of the tryptophan hydroxylase (TPH2) has generated new interest in the connection between serotonergic systems and depression [39]. The TPH2 gene is located on chromosome 12q, a region previously implicated in linkage studies of BPD.Zill et al. [40] reported the first evidence of an association of variants in the THP2 gene and major depression. Zhang et al. [41] identified a functional polymorphism (Arg441His) that results in about 80% loss of function in serotonin production when expressed in a cell system. The authors also reported that this rare mutation was not seen in 219 healthy controls but was seen in 9 of 87 individuals with major depression [41]. However, subsequent replication attempts by other groups for this rare variant were negative. Haplotypic associations of sets of markers across the TPH2 gene have yielded positive results [42] and, interestingly, variants were associated with suicidal behavior [43, 44]. Deficits in brain serotonin synthesis secondary to genetic variation in the TPH2 gene might represent an important risk factor for unipolar depression.In light of these multiple positive and negative reports for various candidate genes, several recent meta-analyses have tried to dissect the genetic factors of depression. The hope was that by combining several underpowered small studies into one large study, the meta-analysis would have sufficient power to detect risk alleles. One of the most comprehensive meta-analyses in depression was recently published and included 183 articles covering 393 polymorphisms in 102 genes [45]. The study showed a potential role for variants in the genes APOE (apolipoprotein E), GNB3 (guanine nucleotide-binding protein β-3), MTHFR (methylene tetrahydrofolate reductase), and SLC6A4 (serotonin transporter). Lopez-Leon et al. [45] observed that the S allele of the 5-HTTLPRconferred a small increase of risk of MDD, with an OR of 1.11; however, another meta-analytic study focusing only on the 5-HTTLPRvariant recently yielded no evidence that the serotonin transporter genotype alone or in interaction with stressful life events is associated with an elevated risk of depression in men alone, women alone, or both sexes combined [22•]. Although several limitations exist in the interpretation of genetic meta-analyses (eg, publication bias and which studies are included and excluded), the overall picture is that no strong susceptibility gene or set of genetic markers for MDD could be identified.Genome-Wide Association StudiesWith the rapid development of technological advances in genomics, it is now possible to genotype 500,000 to 1 million SNPs across the genome in cases and healthy controls. This GWAS design has the advantage that no genes are preselected (as is the case in candidate gene studies), and robust findings might identify new pathways involved in mood disorders. Limitations of this approach are the immense amount of data, costs, and issues regarding multiple testing [46]. The stringent statistical correction for multiple testing might mask true signals from genes that confer only modest risk of disease [47, 48]. Currently, the genome-wide significance level is set (P<10−7to P<10−8). Despite these obstacles, the first results in complex medical disorders such as Crohn's disease, diabetes, and rheumatoid arthritis have emerged from GWAS [49], and several GWAS have been published for psychiatric disorders [50].Thus far in depression, five large GWAS have been published [51•, 52–55]. Sullivan et al. [51•] used a semi–community-based sample of 1,738 cases and 1,802 controls with MDD collected through the Genetic Association Information Network and conducted a GWAS of 435,291 SNPs. There was no genome-wide significant finding; however, they found 11 SNPs associated with MDD that are localized to a 167-kb region overlapping the gene piccolo (PCLO), a presynpatic protein also known as aczonin that is important in monoaminergic neurotransmission in the brain. A replication attempt of the initial finding failed to reach the significance threshold in five independent samples (6,079 MDD independent cases, 5,893 controls). Additional studies are needed to investigate the role of PCLO in MDD and central nervous system neurotransmission. The second published GWAS of MDD used a German clinic-based sample of 1,022 recurrent depression cases and 1,000 controls and investigated 494,678 SNPs [53]. Similar to the Sullivan et al. [51•] study, none of the SNPs were statistically significant after correction for multiple testing. The authors then performed a meta-analysis combining their first sample with a second sample of Swiss origin (494 cases, 1,052 controls) and found their best signal at rs4238010, 260 kb from the closest gene (CCND2, or cyclin D2); however, they still did not achieve genome-wide significance. Another recent GWAS of recurrent, early-onset MDD (GenRED) with onset before 31 years of age investigated 671,424 SNPs in 1,020 cases and 1,636 controls [56]. Again, no genome-wide significant evidence for association was observed. The strongest evidence for association was observed on chromosome 18q22.1 (rs17077540) in a region previously implicated in linkage scans in BPD. Recently, an additional GWAS of MDD was published that included the STAR*D sample of 1221 MDD cases [55]. In addition, the authors conducted a meta-analysis using three samples (Genetic Association Information Network, GenRED, STAR*D) with a total of 3,956 cases and 3,428 controls. There were no genome-wide significant findings in the primary analysis or any of the secondary analyses. Some intronic markers in the meta-analysis were close to genome-wide significance levels (ie, P<10−6) and included the genes ATP6V1B2, GRM7, and SP4. The SNP in the ATP6V1B2 gene is in close proximity to the adjacent gene VMAT1(vesicular monoamine transporter 1), previously implicated in BPD [56]. GRM7 encodes a cell surface receptor and might be an interesting new target for drug development. Interestingly, weak signals were also observed by Muglia et al. [53] and Sullivan and colleagues [51•] for this gene. The gene SP4 encodes a brain-specific zinc finger transcription factor, and several studies have shown an association between SP4and an SP4-binding site in GRK3 (G-protein receptor kinase 3) and BPD [57]. The most recently published GWAS of MDD by Lewis et al. [52] also failed to identify genome-wide significant genetic variants contributing to major depression. The authors included 1636 cases of depression ascertained in the United Kingdom and 1594 controls. One SNP in the BICC1 gene achieved suggestive evidence for association. A meta-analysis of United Kingdom data with previously published results from Muglia et al [53] showed some evidence for association near neuroligin-1 (NLGN1) on chromosome 3 but did not support findings at BICC1.Although the results of these five GWAS in MDD were all essentially negative, they do suggest interesting candidate genes that may be worthwhile to follow up in future studies. It is becoming increasingly clear that individual genetic susceptibility factors for depression are likely to have only minor effects, and very large pooled analyses of cases and controls will be necessary to identify them [50]. One of the first attempts of these large-scale analyses recently yielded interesting results. A meta-analysis that used GWAS data from BPD and MDD cohorts, including more than 13,600 individuals, identified six SNPs at 3p21.1 associated with major mood disorders (rs2251219; P = 3.63 × 10−8; OR, 0.87) [58•]. Supportive evidence for association was observed in two of three independent replication cohorts. These results provide an example of a shared genetic susceptibility locus for BPD and MDD and point out that perhaps our current dichotomous conceptualization of mood disorders is incorrect.Conclusions and Future DirectionsThe field of psychiatric genetics in general has been disappointing given that the initial hope to find common gene variants of large effect in the pathogenesis of mental illnesses has been unsuccessful. In most psychiatric illnesses, the phenotype seems too complex, with the patient cohorts too small, and no findings have been consistently replicated. This is also the case for MDDs. In addition, the phenotypic effects of genetic variants identified to date are weak, with ORs of 1.0 to 1.2. The picture is further complicated when comparing the magnitude of the impact of gene variation on disease susceptibility with the impact of lifestyle and environmental factors, which is likely to be large [59]. Despite these obstacles, the field of psychiatric genetics is rapidly growing, and several new technological advances (eg, whole-genome sequencing) will be soon available for large-scale studies. It is important to remember that genetic information will only provide additional information on one aspect of the complex and personal history of psychiatric patients. It is the sum of inside and outside factors that contributes and influences mental pathology and well-being.FootnotesDisclosure Dr. Lohoff reported no potential conflict of interest relevant to this article.Article informationCurr Psychiatry Rep. Author manuscript; available in PMC 2011 Dec 1.Published in final edited form as:Curr Psychiatry Rep. 2010 Dec; 12(6): 539–546.doi: 10.1007/s11920-010-0150-6PMCID: PMC3077049NIHMSID: NIHMS254781PMID: 20848240Falk W. LohoffFalk W. Lohoff, Department of Psychiatry, Translational Research Laboratory, University of Pennsylvania School of Medicine, Center for Neurobiology and Behavior, 125 South 31st Street, Room 2213, Philadelphia, PA 19104, USA;Corresponding author.Falk W. Lohoff: ude.nnepu.dem.liam@ffoholCopyright noticeThe publisher's final edited version of this article is available at Curr Psychiatry RepSee other articles in PMC that cite the published article.ReferencesPapers of particular interest, published recently, have been highlighted as:• Of importance1. Tsuang MT, Taylor L, Faraone SV. An overview of the genetics of psychotic mood disorders. J Psychiatr Res. 2004;38:3–15. [PubMed]2. Kessler RC, Chiu WT, Demler O, et al. Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62:617–627. [PMC free article][PubMed]3. Lohoff FW, Berrettini WH. Genetics of mood disorders. In: Charney DS, editor. Neurobiology of Mental Illness.New York: Oxford University Press; 2008. p. 1504.4. Sullivan PF, Neale MC, Kendler KS. Genetic epidemiology of major depression: review and meta-analysis. Am J Psychiatry. 2000;157:1552–1562.[PubMed]5. Weissman MM, Wickramaratne P, Adams PB, et al. The relationship between panic disorder and major depression. A new family study. Arch Gen Psychiatry. 1993;50:767–780.[PubMed]6. Levinson DF. The genetics of depression: a review. Biol Psychiatry. 2006;60:84–92. [PubMed]7. Holmans P, Zubenko GS, Crowe RR, et al. Genomewide significant linkage to recurrent, early-onset major depressive disorder on chromosome 15q. Am J Hum Genet. 2004;74:1154–1167. [PMC free article] [PubMed]8. Holmans P, Weissman MM, Zubenko GS, et al. Genetics of recurrent early-onset major depression (GenRED): final genome scan report. Am J Psychiatry. 2007;164:248–258. [PubMed]9. Levinson DF, Evgrafov OV, Knowles JA, et al. Genetics of recurrent early-onset major depression (GenRED): significant linkage on chromosome 15q25-q26 after fine mapping with single nucleotide polymorphism markers. Am J Psychiatry. 2007;164:259–264. [PubMed]10. Abkevich V, Camp NJ, Hensel CH, et al. Predisposition locus for major depression at chromosome 12q22-12q23.2. Am J Hum Genet. 2003;73:1271–1281. [PMC free article][PubMed]11. Camp NJ, Lowry MR, Richards RL, et al. Genome-wide linkage analyses of extended Utah pedigrees identifies loci that influence recurrent, early-onset major depression and anxiety disorders. Am J Med Genet B Neuropsychiatr Genet. 2005;135:85–93.[PubMed]12. McGuffin P, Knight J, Breen G, et al. Whole genome linkage scan of recurrent depressive disorder from the Depression Network study. Hum Mol Genet. 2005;14:3337–3345. [PubMed]13. Hamilton SP, Fyer AJ, Durner M, et al. Further genetic evidence for a panic disorder syndrome mapping to chromosome 13q. Proc Natl Acad Sci U S A. 2003;100:2550–2555.[PMC free article] [PubMed]14. Middeldorp CM, Sullivan PF, Wray NR, et al. Suggestive linkage on chromosome 2, 8, and 17 for lifetime major depression. Am J Med Genet B Neuropsychiatr Genet. 2009;150B:352–358. [PMC free article] [PubMed]15. Cloninger CR, Van Eerdewegh P, Goate A, et al. Anxiety proneness linked to epistatic loci in genome scan of human personality traits. Am J Med Genet. 1998;81:313–317. [PubMed]16. Fullerton J, Cubin M, Tiwari H, et al. Linkage analysis of extremely discordant and concordant sibling pairs identifies quantitative-trait loci that influence variation in the human personality trait neuroticism. Am J Hum Genet. 2003;72:879–890.[PMC free article] [PubMed]17. Caspi A, Hariri AR, Holmes A, et al. Genetic sensitivity to the environment: the case of the serotonin transporter gene and its implications for studying complex diseases and traits. Am J Psychiatry. 2010;167:509–527.[PMC free article] [PubMed]18. Goldman N, Glei DA, Lin YH, et al. The serotonin transporter polymorphism (5-HTTLPR): allelic variation and links with depressive symptoms. Depress Anxiety. 2010;27:260–269. [PMC free article][PubMed]19. Uher R, McGuffin P. The moderation by the serotonin transporter gene of environmental adversity in the etiology of depression: 2009 update. Mol Psychiatry. 2010;15:18–22.[PubMed]20. Lesch KP, Bengel D, Heils A, et al. Association of anxiety-related traits with a polymorphism in the serotonin transporter gene regulatory region. Science. 1996;274:1527–1531. [PubMed]21. Caspi A, Sugden K, Moffitt TE, et al. Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science. 2003;301:386–389.[PubMed]22•. Risch N, Herrell R, Lehner T, et al. Interaction between the serotonin transporter gene (5-HTTLPR), stressful life events, and risk of depression: a meta-analysis. JAMA. 2009;301:2462–2471. [PMC free article] [PubMed]This large meta-analysis of the serotonin transporter gene 5-HTTLPRpolymorphism found no evidence for an association with stressful life events or increased risk for depression.23. Anguelova M, Benkelfat C, Turecki G. A systematic review of association studies investigating genes coding for serotonin receptors and the serotonin transporter: I. Affective disorders. Mol Psychiatry. 2003;8:574–591. [PubMed]24. Lohoff FW, Ferraro TN. Pharmacogenetic considerations in the treatment of psychiatric disorders. Expert Opin Pharmacother. 2010;11:423–439. [PubMed]25. Serretti A, Kato M, De Ronchi D, et al. Meta-analysis of serotonin transporter gene promoter polymorphism (5-HTTLPR) association with selective serotonin reuptake inhibitor efficacy in depressed patients. Mol Psychiatry. 2007;12:247–257. [PubMed]26. McMahon FJ, Buervenich S, Charney D, et al. Variation in the gene encoding the serotonin 2A receptor is associated with outcome of antidepressant treatment. Am J Hum Genet. 2006;78:804–814.[PMC free article] [PubMed]27. Uher R, Farmer A, Maier W, et al. Measuring depression: comparison and integration of three scales in the GENDEP study. Psychol Med. 2008;38:289–300. [PubMed]28. Uher R, Huezo-Diaz P, Perroud N, et al. Genetic predictors of response to antidepressants in the GENDEP project. Pharmacogenomics J. 2009;9:225–233.[PubMed]29. Kendler KS, Kessler RC, Walters EE, et al. Stressful life events, genetic liability, and onset of an episode of major depression in women. Am J Psychiatry. 1995;152:833–842.[PubMed]30. Jokela M, Keltikangas-Jarvinen L, Kivimaki M, et al. Serotonin receptor 2A gene and the influence of childhood maternal nurturance on adulthood depressive symptoms. Arch Gen Psychiatry. 2007;64:356–360. [PubMed]31. Castren E, Rantamaki T. The role of BDNF and its receptors in depression and antidepressant drug action: reactivation of developmental plasticity. Dev Neurobiol. 2010;70:289–297. [PubMed]32. Post RM. Role of BDNF in bipolar and unipolar disorder: clinical and theoretical implications. J Psychiatr Res. 2007;41:979–990. [PubMed]33. Duman RS. Role of neurotrophic factors in the etiology and treatment of mood disorders. Neuromolecular Med. 2004;5:11–25. [PubMed]34. Roceri M, Cirulli F, Pessina C, et al. Postnatal repeated maternal deprivation produces age-dependent changes of brain-derived neurotrophic factor expression in selected rat brain regions. Biol Psychiatry. 2004;55:708–714. [PubMed]35. Nibuya M, Morinobu S, Duman RS. Regulation of BDNF and trkB mRNA in rat brain by chronic electroconvulsive seizure and antidepressant drug treatments. J Neurosci. 1995;15:7539–7547. [PubMed]36. Schumacher J, Jamra RA, Becker T, et al. Evidence for a relationship between genetic variants at the brain-derived neurotrophic factor (BDNF) locus and major depression. Biol Psychiatry. 2005;58:307–314. [PubMed]37. Surtees PG, Wainwright NW, Willis-Owen SA, et al. No association between the BDNF Val66Met polymorphism and mood status in a non-clinical community sample of 7389 older adults. J Psychiatr Res. 2007;41:404–409. [PubMed]38. Hashimoto K. Brain-derived neurotrophic factor as a biomarker for mood disorders: an historical overview and future directions. Psychiatry Clin Neurosci. 2010;64:341–357. [PubMed]39. Walther DJ, Bader M. A unique central tryptophan hydroxylase isoform. Biochem Pharmacol. 2003;66:1673–1680. [PubMed]40. Zill P, Baghai TC, Zwanzger P, et al. SNP and haplotype analysis of a novel tryptophan hydroxylase isoform (TPH2) gene provide evidence for association with major depression. Mol Psychiatry. 2004;9:1030–1036.[PubMed]41. Zhang X, Gainetdinov RR, Beaulieu JM, et al. Loss-of-function mutation in tryptophan hydroxylase-2 identified in unipolar major depression. Neuron. 2005;45:11–16. [PubMed]42. Van Den Bogaert A, Sleegers K, De Zutter S, et al. Association of brain-specific tryptophan hydroxylase, TPH2, with unipolar and bipolar disorder in a Northern Swedish, isolated population. Arch Gen Psychiatry. 2006;63:1103–1110. [PubMed]43. Lopez VA, Detera-Wadleigh S, Cardona I, et al. Nested association between genetic variation in tryptophan hydroxylase II, bipolar affective disorder, and suicide attempts. Biol Psychiatry. 2007;61:181–186. [PubMed]44. De Luca V, Mueller DJ, Tharmalingam S, et al. Analysis of the novel TPH2 gene in bipolar disorder and suicidality. Mol Psychiatry. 2004;9:896–897. [PubMed]45. Lopez-Leon S, Janssens AC, Gonzalez-Zuloeta Ladd AM, et al. Meta-analyses of genetic studies on major depressive disorder. Mol Psychiatry. 2008;13:772–785. [PubMed]46. Pearson TA, Manolio TA. How to interpret a genome-wide association study. JAMA. 2008;299:1335–1344.[PubMed]47. Jorgenson E, Witte JS. A gene-centric approach to genome-wide association studies. Nat Rev Genet. 2006;7:885–891. [PubMed]48. Clark AG, Boerwinkle E, Hixson J, et al. Determinants of the success of whole-genome association testing. Genome Res. 2005;15:1463–1467.[PubMed]49. Welcome Trust Case Control Consortium Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature. 2007;447:661–678.[PMC free article] [PubMed]50. Cichon S, Craddock N, Daly M, et al. Genomewide association studies: history, rationale, and prospects for psychiatric disorders. Am J Psychiatry. 2009;166:540–556. [PMC free article][PubMed]51•. Sullivan PF, de Geus EJ, Willemsen G, et al. Genome-wide association for major depressive disorder: a possible role for the presynaptic protein piccolo. Mol Psychiatry. 2009;14:359–375. [PMC free article] [PubMed]This was the first GWAS in depression. Results did not achieve genome-wide significance but suggest a potential role for the PCLO gene.52. Lewis CM, Ng MY, Butler AW, et al. Genome-wide association study of major recurrent depression in the U.K. population. Am J Psychiatry. 2010;167:949–957. [PubMed]53. Muglia P, Tozzi F, Galwey NW, et al. Genome-wide association study of recurrent major depressive disorder in two European case-control cohorts. Mol Psychiatry. 2010;15:589–601.[PubMed]54. Shi J, Potash JB, Knowles JA, et al. Genome-wide association study of recurrent early-onset major depressive disorder. Mol Psychiatry. 2010 Feb 2;Epub ahead of print. [PubMed]55. Shyn SI, Shi J, Kraft JB, et al. Novel loci for major depression identified by genome-wide association study of Sequenced Treatment Alternatives to Relieve Depression and meta-analysis of three studies. Mol Psychiatry. 2009 Dec 29; Epub ahead of print. [PMC free article] [PubMed]56. Lohoff FW, Dahl JP, Ferraro TN, et al. Variations in the vesicular monoamine transporter 1 gene (VMAT1/SLC18A1) are associated with bipolar I disorder. Neuropsychopharmacology. 2006;31:2739–2747. [PMC free article][PubMed]57. Zhou X, Tang W, Greenwood TA, et al. Transcription factor SP4 is a susceptibility gene for bipolar disorder. PLoS One. 2009;4:e5196.[PMC free article] [PubMed]58•. McMahon FJ, Akula N, Schulze TG, et al. Bipolar Disorder Genome Study (BiGS) Consortium Meta-analysis of genome-wide association data identifies a risk locus for major mood disorders on 3p21.1. Nat Genet. 2010;42:128–131. [PMC free article][PubMed]This is the largest meta-analysis of mood disorders to date, including more than 13,600 individuals genotyped on high-density SNP arrays. The study identified SNPs at 3p21.1 associated with major mood disorders.59. Willett WC. Balancing life-style and genomics research for disease prevention. Science. 2002;296:695–698.[PubMed]

What are the effects of long term adderall use?

Therapeutic PO doses of L and D amphetamine do not pose any significant long term risks if taken as directed for the appropriate condition in terms of the literature I have read in peer reviewed studies from academic journals:Potential Adverse Effects of Amphetamine Treatment on Brain and Behavior: A Review==========================================================I have been on methylphenidate, Adderral, Dexedrine and lisdexamphetamine for over twenty years. (separately) and while my experience is entirely anecdotal with a pretty small sample size and no control………The benefits of being able to function outweigh any potential long term risks for me.My concern when I was put on both methylphenidate and Mirapex (a dopamine agonist) together whether the increase in dopamine activity might trigger Parkinson’s or some other horrible issue. I was assured there was no correlation in my case.So do a risk reward matrix and figure out the best choice with your physician, Be an active challenging , questioning, patientIt is your right!1. Volkow ND, Insel TR. What are the long-term effects of methylphenidate treatment? Biol Psychiatry. 2003 Dec 15;54(12):1307–1309. [PubMed] [Google Scholar]2. Fleckenstein AE, Volz TJ, Riddle EL, Gibb JW, Hanson GR. New insights into the mechanism of action of amphetamines. Annual Review of Pharmacology and Toxicology. 2007;47:681–698. [PubMed] [Google Scholar]3. Clement BA, Goff CM, Forbes TDA. Toxic amines and alkaloids from Acacia berlandieri. Phytochemistry. 1997;46(2):249–254. [Google Scholar]4. Clement BA, Goff CM, Forbes TDA. Toxic amines and alkaloids from Acacia rigidula. Phytochemistry. 1998;49(5):1377–1380. [Google Scholar]5. Goodman LS, Hardman JG, Limbird LE, Gilman AG. Goodman & Gilman's The Pharmacological Basis of Therapeutics. McGraw-Hill; New York: 2001. [Google Scholar]6. Madras BK, Miller GM, Fischman AJ. The dopamine transporter and attention-deficit/hyperactivity disorder. Biological Psychiatry. 2005;57(11):1397–1409. [PubMed] [Google Scholar]7. Boutrel B, Koob GF. What keeps us awake: the neuropharmacology of stimulants and wakefulness-promoting medications. Sleep. 2004;27(6):1181–1194. [PubMed] [Google Scholar]8. Elia J, Borcherding BG, Potter WZ, Mefford IN, Rapoport JL, Keysor CS. Stimulant drug treatment of hyperactivity: biochemical correlates. Clinical Pharmacology and Therapeutics. 1990;48(1):57–66. [PubMed] [Google Scholar]9. Kuczenski R, Segal DS, Cho AK, Melega W. Hippocampus norepinephrine, caudate dopamine and serotonin, and behavioral responses to the stereoisomers of amphetamine and methamphetamine. Journal of Neuroscience. 1995;15(2):1308–1317. [PMC free article] [PubMed] [Google Scholar]10. Bradley C. The behavior of children receiving benzedrine. American Journal of Psychiatry. 1937;94:577–585. [Google Scholar]11. International Narcotics Control Board Staff . Psychotropic Substances. United Nations Publications; 2004. [Google Scholar]12. Education USDo . Identifying and Treating Attention Deficit Hyperactivity Disorder: A Resource for School and Home. U.S. Department of Education, Office of Special Education and Rehabilitative Services, Office of Special Education Programs; Washington, D.C.: 2003. [Google Scholar]13. Zametkin AJ, Nordahl TE, Gross M, King AC, Semple WE, Rumsey J, et al. Cerebral glucose metabolism in adults with hyperactivity of childhood onset. New England Journal of Medicine. 1990;323(20):1361–1366. [PubMed] [Google Scholar]14. Goldman LS, Genel M, Bezman RJ, Slanetz PJ. Diagnosis and treatment of attention-deficit/hyperactivity disorder in children and adolescents. Council on Scientific Affairs, American Medical Association. Journal of the American Medical Association. 1998;279(14):1100–1107. [PubMed] [Google Scholar]15. McGough JJ, Biederman J, Greenhill LL, McCracken JT, Spencer TJ, Posner K, et al. Pharmacokinetics of SLI381 (ADDERALL XR), an extended-release formulation of Adderall. Journal of American Academy of Child Acolescent Psychiatry. 2003;42(6):684–691. [PubMed] [Google Scholar]16. Goodman DW. Lisdexamfetamine Dimesylate: The First Prodrug Stimulant. Psychiatry MMC. 2007 [PMC free article] [PubMed] [Google Scholar]17. Adler L. American Academy of Child and Adolescent Psychiatry. Boston, MA: 2007. Efficacy and Safety of Lisdexamfetamine Dimesylate in Adults with Attention Deficit Hyperactvity Disorder. al. e. [Google Scholar]18. Connor DF, Steingard RJ. New formulations of stimulants for attention-deficit hyperactivity disorder: therapeutic potential. CNSDrugs. 2004;18(14):1011–1030. [PubMed] [Google Scholar]19. Wilens TE. Drug therapy for adults with attention-deficit hyperactivity disorder. Drugs. 2003;63(22):2395–2411. [PubMed] [Google Scholar]20. Wilens TE, Faraone SV, Biederman J. Attention-deficit/hyperactivity disorder in adults. Journal of the American Medical Association. 2004;292(5):619–623. [PubMed] [Google Scholar]21. Mitler MM, Aldrich MS, Koob GF, Zarcone VP. Narcolepsy and its treatment with stimulants. ASDA standards of practice. Sleep. 1994;17(4):352–371. [PubMed] [Google Scholar]22. Mitler MM, Hajdukovic R. Relative efficacy of drugs for the treatment of sleepiness in narcolepsy. Sleep. 1991;14(3):218–220. [PMC free article] [PubMed] [Google Scholar]23. National Toxicology P NTP-CERHR monograph on the potential human reproductive and developmental effects of amphetamines. 2005. Report No.: 16. [PubMed]24. Clinical practice guideline: treatment of the school-aged child with attention-deficit/hyperactivity disorder. Pediatrics. 2001;108(4):1033–1044. [PubMed] [Google Scholar]25. Efron D, Jarman F, Barker M. Side effects of methylphenidate and dexamphetamine in children with attention deficit hyperactivity disorder: a double-blind, crossover trial. Pediatrics. 1997;100(4):662–666. [PubMed] [Google Scholar]26. Frishman WH, Del VA, Sanal S, Ismail A. Cardiovascular manifestations of substance abuse: part 2: alcohol, amphetamines, heroin, cannabis, and caffeine. Heart Dis. 2003;5(4):253–271. [PubMed] [Google Scholar]27. Safer DJ, Zito JM, dosReis S. Concomitant psychotropic medication for youths. American Journal of Psychiatry. 2003;160(3):438–449. [PubMed] [Google Scholar]28. dosReis S, Zito JM, Safer DJ, Gardner JF, Puccia KB, Owens PL. Multiple psychotropic medication use for youths: a two-state comparison. JChild AdolescPsychopharmacol. 2005;15(1):68–77. [PubMed] [Google Scholar]29. CDC. Attention-Deficit / Hyperactivity Disorder (ADHD) 2005. [2008/01/23/]. Available from: Attention-Deficit / Hyperactivity Disorder (ADHD) (Attention-Deficit / Hyperactivity Disorder (ADHD))30. Biederman J, Faraone SV. Current concepts on the neurobiology of Attention-Deficit/Hyperactivity Disorder. JAttenDisord. 2002;6(Suppl 1):S7–16. [PubMed] [Google Scholar]31. Faraone SV, Biederman J, Spencer TJ, Aleardi M. Comparing the efficacy of medications for ADHD using meta-analysis. MedGenMed. 2006;8(4):4. [PMC free article] [PubMed] [Google Scholar]32. Faber A, Kalverdijk LJ, de Jong-van den Berg LT, Hugtenburg JG, Minderaa RB, Tobi H. Parents report on stimulant-treated children in the Netherlands: initiation of treatment and follow-up care. JChild AdolescPsychopharmacol. 2006;16(4):432–440. [PubMed] [Google Scholar]33. Preen DB, Calver J, Sanfilippo FM, Bulsara M, Holman CD. Patterns of psychostimulant prescribing to children with ADHD in Western Australia: variations in age, gender, medication type and dose prescribed. AustNZJPublic Health. 2007;31(2):120–126. [PubMed] [Google Scholar]34. Sankaranarayanan J, Puumala SE, Kratochvil CJ. Diagnosis and treatment of adult attention-deficit/hyperactivity disorder at US ambulatory care visits from 1996 to 2003. CurrMedResOpin. 2006;22(8):1475–1491. [PubMed] [Google Scholar]35. Wu EQ, Birnbaum HG, Zhang HF, Ivanova JI, Yang E, Mallet D. Health care costs of adults treated for attention-deficit/hyperactivity disorder who received alternative drug therapies. JManagCare Pharm. 2007;13(7):561–569. [PubMed] [Google Scholar]36. Brown GL, Hunt RD, Ebert MH, Bunney WE, Jr., Kopin IJ. Plasma levels of d-amphetamine in hyperactive children. Serial behavior and motor responses. Psychopharmacology (Berlin) 1979;62(2):133–140. [PubMed] [Google Scholar]37. Thorpy M. Therapeutic advances in narcolepsy. Sleep Med. 2007;8(4):427–440. [PubMed] [Google Scholar]38. Dowling GJ, Weiss SR, Condon TP. Drugs of abuse and the aging brain. Neuropsychopharmacology. 2008;33(2):209–218. [PubMed] [Google Scholar]39. Substance A, Mental Health Services A. Detailed Emergency Department Tables from DAWN: 2002. 2002. [2008/01/23/]. Available from: http://dawninfo.samhsa.gov/old_dawn/pubs_94_02/pickatable/2001/2.8.0.xls (http://dawninfo.samhsa.gov/old_dawn/pubs_94_02/pickatable/2001/2.8.0.xls).40. Gfroerer J, Penne M, Pemberton M, Folsom R. Substance abuse treatment need among older adults in 2020: the impact of the aging baby-boom cohort. Drug and Alcohol Dependence. 2003;69(2):127–135. [PubMed] [Google Scholar]41. Bowyer JF, Holson RR, Chang LW, Dyer RS. Handbook of Neurotoxicology. Marcel Dekker, Inc; New York: 1995. Methamphetamine and amphetamine neurotoxicity. pp. 845–870. [Google Scholar]42. Seiden LS, Sabol KE, Chang LW, Dyer RS. Handbook of Neurotoxicology. Marcel Dekker, Inc; New York: 1995. Neurotoxicity of methamphetamine-related drugs and cocaine. pp. 825–843. [Google Scholar]43. Kita T, Wagner GC, Nakashima T. Current research on methamphetamine-induced neurotoxicity: animal models of monoamine disruption. JPharmacolSci. 2003;92(3):178–195. [PubMed] [Google Scholar]44. Selemon LD, Begovic A, Goldman-Rakic PS, Castner SA. Amphetamine sensitization alters dendritic morphology in prefrontal cortical pyramidal neurons in the non-human primate. Neuropsychopharmacology. 2007 Apr;32(4):919–931. [PubMed] [Google Scholar]45. Tata DA, Yamamoto BK. Interactions between methamphetamine and environmental stress: role of oxidative stress, glutamate and mitochondrial dysfunction. Addiction. 2007;102(Suppl 1):49–60. [PubMed] [Google Scholar]46. Segal DS, Kuczenski R. Escalating dose-binge treatment with methylphenidate: role of serotonin in the emergent behavioral profile. JPharmacolExpTher. 1999;291(1):19–30. [PubMed] [Google Scholar]47. Yuan J, McCann U, Ricaurte G. Methylphenidate and brain dopamine neurotoxicity. Brain Research. 1997;767(1):172–175. [PubMed] [Google Scholar]48. Fischer JF, Cho AK. Chemical release of dopamine from striatal homogenates: evidence for an exchange diffusion model. J Pharmacol Exp Ther. 1979 Feb;208(2):203–209. [PubMed] [Google Scholar]49. Krueger BK. Kinetics and block of dopamine uptake in synaptosomes from rat caudate nucleus. Journal of Neurochemistry. 1990;55:260–267. [PubMed] [Google Scholar]50. Segal DS, Kuczenski R. Repeated binge exposure to amphetamine and methamphetamine: Behavioral and neurochemical characterization. JPharmacolExpTher. 1997;282:561–573. [PubMed] [Google Scholar]51. Melega WP, Raleigh MJ, Stout DB, Lacan G, Huang SC, Phelps ME. Recovery of striatal dopamine function after acute amphetamine- and methamphetamine-induced neurotoxicity in the vervet monkey. Brain Research. 1997;766:113–120. [PubMed] [Google Scholar]52. Moll GH, Hause S, Ruther E, Rothenberger A, Huether G. Early methylphenidate administration to young rats causes a persistent reduction in the density of striatal dopamine transporters. JChild AdolescPsychopharmacol. 2001;11(1):15–24. [PubMed] [Google Scholar]53. Ricaurte GA, Mechan AO, Yuan J, Hatzidimitriou G, Xie T, Mayne AH, et al. Amphetamine treatment similar to that used in the treatment of adult attention-deficit/hyperactivity disorder damages dopaminergic nerve endings in the striatum of adult nonhuman primates. JPharmacolExpTher. 2005;315(1):91–98. [PubMed] [Google Scholar]54. Borcherding BG, Keysor CS, Cooper TB, Rapoport JL. Differential effects of methylphenidate and dextroamphetamine on the motor activity level of hyperactive children. Neuropsychopharmacology. 1989;2(4):255–263. [PubMed] [Google Scholar]55. Greenhill LL, Swanson JM, Steinhoff K, Fried J, Posner K, Lerner M, et al. A pharmacokinetic/pharmacodynamic study comparing a single morning dose of adderall to twice-daily dosing in children with ADHD. Journal of American Academy of Child Acolescent Psychiatry. 2003;42(10):1234–1241. [PubMed] [Google Scholar]56. Anderson LI, Leipheimer RE, Dluzen DE. Effects of neonatal and prepubertal hormonal manipulations upon estrogen neuroprotection of the nigrostriatal dopaminergic system within female and male mice. Neuroscience. 2005;130(2):369–382. [PubMed] [Google Scholar]57. Bowyer JF, Gough B, Slikker W, Jr., Lipe GW, Newport GD, Holson RR. Effects of a cold environment or age on methamphetamine-induced dopamine release in the caudate putamen of female rats. Pharmacology Biochemistry and Behavior. 1993;44(1):87–98. [PubMed] [Google Scholar]58. Miller DB, O'Callaghan JP, Ali SF. Age as a susceptibility factor in the striatal dopaminergic neurotoxicity observed in the mouse following substituted amphetamine exposure. Annals of the New York Academy of Sciences. 2000;914:194–207. [PubMed] [Google Scholar]59. Teuchert-Noodt G, Dawirs RR. Age-related toxicity in prefrontal cortex and caudateputamen complex of gerbils (Meriones unguiculatus) after a single dose of methamphetamine. Neuropharmacology. 1991;30(7):733–743. [PubMed] [Google Scholar]60. Truex LL, Schmidt MJ. 3H-amphetamine concentrations in the brains of young and aged rats: implications for assessment of drug effects in aged animals. Neurobiology of Aging. 1980;1(1):93–95. [PubMed] [Google Scholar]61. O'Neil ML, Kuczenski R, Segal DS, Cho AK, Lacan G, Melega WP. Escalating dose pretreatment induces pharmacodynamic and not pharmacokinetic tolerance to a subsequent high-dose methamphetamine binge. Synapse. 2006 Nov;60(6):465–473. [PubMed] [Google Scholar]62. Segal DS, Kuczenski R, O'Neil ML, Melega WP, Cho AK. Escalating dose methamphetamine pretreatment alters the behavioral and neurochemical profiles associated with exposure to a high-dose methamphetamine binge. Neuropsychopharmacology. 2003;28:1730–1740. [PubMed] [Google Scholar]63. Carlsson A, Winblad B. Influence of age and time interval between death and autopsy on dopamine and 3-methoxytyramine levels in human basal ganglia. J Neural Transm. 1976;38(3−4):271–276. [PubMed] [Google Scholar]64. Carlsson A. Aging and Brain Neurotransmitters. In: D P, editor. Funkitionsstorurgen des Gehirns im Alter. 1981. pp. 67–81. [Google Scholar]65. Advokat C. Update on amphetamine neurotoxicity and its relevance to the treatment of ADHD. JAttenDisord. 2007;11(1):8–16. [PubMed] [Google Scholar]66. Poulton A. Growth on stimulant medication; clarifying the confusion: a review. ArchDisChild. 2005;90(8):801–806. [PMC free article] [PubMed] [Google Scholar]67. Pliszka SR, Matthews TL, Braslow KJ, Watson MA. Comparative effects of methylphenidate and mixed salts amphetamine on height and weight in children with attention-deficit/hyperactivity disorder. Journal of American Academy of Child Acolescent Psychiatry. 2006;45(5):520–526. [PubMed] [Google Scholar]68. Spencer TJ, Faraone SV, Biederman J, Lerner M, Cooper KM, Zimmerman B. Does prolonged therapy with a long-acting stimulant suppress growth in children with ADHD? Journal of American Academy of Child Acolescent Psychiatry. 2006;45(5):527–537. [PubMed] [Google Scholar]69. Swanson JM, Elliott GR, Greenhill LL, Wigal T, Arnold LE, Vitiello B, et al. Effects of stimulant medication on growth rates across 3 years in the MTA follow-up. Journal of American Academy of Child Acolescent Psychiatry. 2007;46(8):1015–1027. [PubMed] [Google Scholar]70. Charach A, Figueroa M, Chen S, Ickowicz A, Schachar R. Stimulant treatment over 5 years: effects on growth. Journal of American Academy of Child Acolescent Psychiatry. 2006;45(4):415–421. [PubMed] [Google Scholar]71. Swanson J, Greenhill L, Wigal T, Kollins S, Stehli A, Davies M, et al. Stimulant-related reductions of growth rates in the PATS. Journal of American Academy of Child Acolescent Psychiatry. 2006;45(11):1304–1313. [PubMed] [Google Scholar]72. Srisurapanont M, Jarusuraisin N, Kittirattanapaiboon P. Treatment for amphetamine dependence and abuse. CochraneDatabaseSystRev. 2001;(4):CD003022. [PubMed] [Google Scholar]73. Hill KP, Sofuoglu M. Biological treatments for amfetamine dependence : recent progress. CNSDrugs. 2007;21(10):851–869. [PubMed] [Google Scholar]74. Executive Board AAoP Use of d-amphetamine and related central nervous system stimulants in children. Pediatrics. 1973;51(2):302–305. [PubMed] [Google Scholar]75. Johnston LD, O'Malley PM, Bachman JG, Schulenberg JE. Monitoring the future national survey results on drug use, 1975−2006: Volume I, Secondary school students. National Institute on Drug Abuse; Bethesda, MD: 2007. [Google Scholar]76. Rawson RA, Condon TP. Why do we need an Addiction supplement focused on methamphetamine? Addiction. 2007;102(Suppl 1):1–4. [PubMed] [Google Scholar]77. Sato M, Hida M, Nagase H. Analysis of pyrolysis products of methamphetamine. Journal of Analytical Toxicology. 2004;28(8):638–643. [PubMed] [Google Scholar]78. Musser CJ, Ahmann PA, Theye FW, Mundt P, Broste SK, Mueller-Rizner N. Stimulant use and the potential for abuse in Wisconsin as reported by school administrators and longitudinally followed children. JDevBehavPediatr. 1998;19(3):187–192. [PubMed] [Google Scholar]79. McCabe SE, Teter CJ, Boyd CJ. The use, misuse and diversion of prescription stimulants among middle and high school students. SubstUseMisuse. 2004;39(7):1095–1116. [PubMed] [Google Scholar]80. McCabe SE, Teter CJ, Boyd CJ. Medical use, illicit use and diversion of prescription stimulant medication. Journal of Psychoactive Drugs. 2006;38(1):43–56. [PMC free article] [PubMed] [Google Scholar]81. McCabe SE, Teter CJ, Boyd CJ. Medical use, illicit use, and diversion of abusable prescription drugs. JAmCollHealth. 2006;54(5):269–278. [PMC free article] [PubMed] [Google Scholar]82. Poulin C. Medical and nonmedical stimulant use among adolescents: from sanctioned to unsanctioned use. CMAJ. 2001;165(8):1039–1044. [PMC free article] [PubMed] [Google Scholar]83. Farris KB, McCarthy AM, Kelly MW, Clay D, Gross JN. Issues of medication administration and control in Iowa schools. JSch Health. 2003;73(9):331–337. [PubMed] [Google Scholar]84. Reiterman T. Prescriptions supplanting illegal substances as drugs of choice. Los Angeles Times. 2008 May 18; 2008. [Google Scholar]85. Bassetti C, Aldrich MS. Narcolepsy. Neurologic Clinics. 1996;14(3):545–571. [PubMed] [Google Scholar]86. Stoops WW, Glaser PE, Fillmore MT, Rush CR. Reinforcing, subject-rated, performance and physiological effects of methylphenidate and d-amphetamine in stimulant abusing humans. J Psychopharmacol. 2004 Dec;18(4):534–543. [PubMed] [Google Scholar]87. Kollins SH. Comparing the abuse potential of methylphenidate versus other stimulants: a review of available evidence and relevance to the ADHD patient. J Clin Psychiatry. 2003;64(Suppl 11):14–18. [PubMed] [Google Scholar]88. Stoops WW, Lile JA, Glaser PE, Rush CR. Discriminative stimulus and self-reported effects of methylphenidate, d-amphetamine, and triazolam in methylphenidate-trained humans. Exp Clin Psychopharmacol. 2005 Feb;13(1):56–64. [PubMed] [Google Scholar]89. Spencer TJ, Biederman J, Ciccone PE, Madras BK, Dougherty DD, Bonab AA, et al. PET study examining pharmacokinetics, detection and likeability, and dopamine transporter receptor occupancy of short- and long-acting oral methylphenidate. American Journal of Psychiatry. 2006;163(3):387–395. [PubMed] [Google Scholar]90. Jasinski D, Krishnan S. U.S. Psychiatric & Mental Health Congress. New Orleans, LA: 2006. A double-blind, randomized, placebo- and active-controlled, 6-period crossover study to evaluate the likability, safety, and abuse potential of lisdexamfetamine dimesylate (LDX) in adult stimulant abusers. [Google Scholar]91. Jasinski D, Krishnan S. U.S. Psychiatric & Mental Health Congress. New Orleans, LA: 2006. Abuse liability of intravenous lisdexamfetamine dimesylate (LDX; NRP104). [Google Scholar]92. Wilens TE, Fusillo S. When ADHD and substance use disorders intersect: relationship and treatment implications. CurrPsychiatry Rep. 2007;9(5):408–414. [PubMed] [Google Scholar]93. Wilens TE, Adamson J, Sgambati S, Whitley J, Santry A, Monuteaux MC, et al. Do individuals with ADHD self-medicate with cigarettes and substances of abuse? Results from a controlled family study of ADHD. The American Journal on Addictions. 2007;16(Suppl 1):14–21. [PubMed] [Google Scholar]94. Lambert NM, Hartsough CS. Prospective study of tobacco smoking and substance dependencies among samples of ADHD and non-ADHD participants. JLearnDisabil. 1998;31(6):533–544. [PubMed] [Google Scholar]95. Lambert NM, McLeod M, Schenk S. Subjective responses to initial experience with cocaine: an exploration of the incentive-sensitization theory of drug abuse. Addiction. 2006 May;101(5):713–725. [PubMed] [Google Scholar]96. Kollins SH, MacDonald EK, Rush CR. Assessing the abuse potential of methylphenidate in nonhuman and human subjects: a review. Pharmacol Biochem Behav. 2001 Mar;68(3):611–627. [PubMed] [Google Scholar]97. Vitiello B. Long-term effects of stimulant medications on the brain: possible relevance to the treatment of attention deficit hyperactivity disorder. J Child Adolesc Psychopharmacol. 2001;11(1):25–34. Spring. [PubMed] [Google Scholar]98. Wilens TE, Faraone SV, Biederman J, Gunawardene S. Does stimulant therapy of attention-deficit/hyperactivity disorder beget later substance abuse? A meta-analytic review of the literature. Pediatrics. 2003;111(1):179–185. [PubMed] [Google Scholar]99. Kuczenski R, Segal DS. Exposure of adolescent rats to oral methylphenidate: preferential effects on extracellular norepinephrine and absence of sensitization and cross-sensitization to methamphetamine. J Neurosci. 2002 Aug 15;22(16):7264–7271. [PMC free article] [PubMed] [Google Scholar]100. Andersen MB, Fuxe K, Werge T, Gerlach J. The adenosine A2A receptor agonist CGS 21680 exhibits antipsychotic-like activity in Cebus apella monkeys. Behav Pharmacol. 2002 Dec;13(8):639–644. [PubMed] [Google Scholar]101. Mague SD, Andersen SL, Carlezon WA., Jr Early developmental exposure to methylphenidate reduces cocaine-induced potentiation of brain stimulation reward in rats. Biol Psychiatry. 2005 Jan 15;57(2):120–125. [PubMed] [Google Scholar]102. Brandon CL, Marinelli M, Baker LK, White FJ. Enhanced reactivity and vulnerability to cocaine following methylphenidate treatment in adolescent rats. Neuropsychopharmacology. 2001 Nov;25(5):651–661. [PubMed] [Google Scholar]103. Mannuzza S, Klein RG, Truong NL, Moulton JL, 3rd, Roizen ER, Howell KH, et al. Age of methylphenidate treatment initiation in children with ADHD and later substance abuse: prospective follow-up into adulthood. Am J Psychiatry. 2008 May;165(5):604–609. [PMC free article] [PubMed] [Google Scholar]104. Spear LP. The adolescent brain and age-related behavioral manifestations. NeurosciBiobehavRev. 2000;24(4):417–463. [PubMed] [Google Scholar]105. Holland PC, Gallagher M. Amygdala circuitry in attentional and representational processes. Trends Cogn Sci. 1999 Feb;3(2):65–73. [PubMed] [Google Scholar]106. Frantz K, Van Hartesveldt C. The locomotor effects of MK801 in the nucleus accumbens of developing and adult rats. Eur J Pharmacol. 1999 Mar 5;368(2−3):125–135. [PubMed] [Google Scholar]107. Teicher MHA, S.L Limbic serotonin turnover plunges during puberty.. Society for Neuroscience Annual Conference; Miami Beach, FL. 1999. [Google Scholar]108. Kalsbeek A, Voorn P, Buijs RM, Pool CW, Uylings HB. Development of the dopaminergic innervation in the prefrontal cortex of the rat. J Comp Neurol. 1988 Mar 1;269(1):58–72. [PubMed] [Google Scholar]109. Leslie CA, Robertson MW, Cutler AJ, Bennett JP., Jr. Postnatal development of D1 dopamine receptors in the medial prefrontal cortex, striatum and nucleus accumbens of normal and neonatal 6-hydroxydopamine treated rats: a quantitative autoradiographic analysis. Brain Res Dev Brain Res. 1991 Sep 19;62(1):109–114. [PubMed] [Google Scholar]110. van Eden CGK, J.M., Uylings HBM. The development of the rat prefrontal cortex: Its size and development of connections with thalamus, spinal cord and other cortical areas. In: Uylings HBMvE, C.G., De Bruin JPC, Corner MA, Feenstra MGP., editors. Progress in brain research, The prefrontal cortex: its structure, function and pathology. Elsevier; Amsterdam: 1990. pp. 169–183. [PubMed] [Google Scholar]111. Kellogg CK, Awatramani GB, Piekut DT. Adolescent development alters stressor-induced Fos immunoreactivity in rat brain. Neuroscience. 1998 Apr;83(3):681–689. [PubMed] [Google Scholar]112. Heimer LdO, J., Alheid GF, Zaborszky L. Perestroika in the basal forebrain: Opening the border between neurology and psychiatry. In: G H, editor. Progress in brain research, Role of the forebrain in sensation and behavior. Vol. 87. Elsevier; Amsterdam: 1991. pp. 109–165. [PubMed] [Google Scholar]113. Powell EW, Leman RB. Connections of the nucleus accumbens. Brain Res. 1976 Apr 9;105(3):389–403. [PubMed] [Google Scholar]114. Risold PY, Thompson RH, Swanson LW. The structural organization of connections between hypothalamus and cerebral cortex. Brain Res Brain Res Rev. 1997 Sep 19;24(2−3):197–254. [PubMed] [Google Scholar]115. Saphier D, Feldman S. Effects of neural stimuli on paraventricular nucleus neurones. Brain Res Bull. 1985 May;14(5):401–407. [PubMed] [Google Scholar]116. Bolanos CA, Glatt SJ, Jackson D. Subsensitivity to dopaminergic drugs in periadolescent rats: a behavioral and neurochemical analysis. Brain Res Dev Brain Res. 1998 Nov 1;111(1):25–33. [PubMed] [Google Scholar]117. Lanier LP, Isaacson RL. Early developmental changes in the locomotor response to amphetamine and their relation to hippocampal function. Brain Res. 1977 May 13;126(3):567–575. [PubMed] [Google Scholar]118. Laviola G, Adriani W, Terranova ML, Gerra G. Psychobiological risk factors for vulnerability to psychostimulants in human adolescents and animal models. Neurosci Biobehav Rev. 1999 Nov;23(7):993–1010. [PubMed] [Google Scholar]119. McKinzie DLM, W.J., Murphy JM, Lumeng L, Li T-K. Rat lines selectively bred for alcohol preference: A potential animal model of adolescent alcohol drinking. In: Hannigan JHS, L.P., Spear NE, Goodlett CR, editors. Alcohol and alcoholism: Effects on brain and development. Lawrence Erlbaum Associates; Mahwah, NJ: 1999. pp. 135–160. [Google Scholar]120. Snyder KJ, Katovic NM, Spear LP. Longevity of the expression of behavioral sensitization to cocaine in preweanling rats. Pharmacol Biochem Behav. 1998 Aug;60(4):909–914. [PubMed] [Google Scholar]121. Spear LP, Brick J. Cocaine-induced behavior in the developing rat. Behav Neural Biol. 1979 Aug;26(4):401–415. [PubMed] [Google Scholar]122. Karreman M, Moghaddam B. The prefrontal cortex regulates the basal release of dopamine in the limbic striatum: an effect mediated by ventral tegmental area. J Neurochem. 1996 Feb;66(2):589–598. [PubMed] [Google Scholar]123. Karler R, Calder LD, Thai DK, Bedingfield JB. The role of dopamine and GABA in the frontal cortex of mice in modulating a motor-stimulant effect of amphetamine and cocaine. Pharmacol Biochem Behav. 1998 May;60(1):237–244. [PubMed] [Google Scholar]124. Kolachana BS, Saunders RC, Weinberger DR. Augmentation of prefrontal cortical monoaminergic activity inhibits dopamine release in the caudate nucleus: an in vivo neurochemical assessment in the rhesus monkey. Neuroscience. 1995 Dec;69(3):859–868. [PubMed] [Google Scholar]125. Cappon GD, Vorhees CV. Plasma and brain methamphetamine concentrations in neonatal rats. Neurotoxicol Teratol. 2001 Jan-Feb;23(1):81–88. [PubMed] [Google Scholar]126. Crawford CA, Williams MT, Newman ER, McDougall SA, Vorhees CV. Methamphetamine exposure during the preweanling period causes prolonged changes in dorsal striatal protein kinase A activity, dopamine D2-like binding sites, and dopamine content. Synapse. 2003 Jun 1;48(3):131–137. [PubMed] [Google Scholar]127. Lucot JB, Wagner GC, Schuster CR, Seiden LS. Decreased sensitivity of rat pups to long-lasting dopamine and serotonin depletions produced by methylamphetamine. Brain Res. 1982 Sep 9;247(1):181–183. [PubMed] [Google Scholar]128. Pu C, Vorhees CV. Developmental dissociation of methamphetamine-induced depletion of dopaminergic terminals and astrocyte reaction in rat striatum. Brain Res Dev Brain Res. 1993 Apr 16;72(2):325–328. [PubMed] [Google Scholar]129. Wagner GC, Schuster CR, Seiden LS. Neurochemical consequences following administration of CNS stimulants to the neonatal rat. Pharmacol Biochem Behav. 1981 Jan;14(1):117–119. [PubMed] [Google Scholar]130. Rice D, Barone S., Jr. Critical periods of vulnerability for the developing nervous system: evidence from humans and animal models. Environ Health Perspect. 2000 Jun;108(Suppl 3):511–533. [PMC free article] [PubMed] [Google Scholar]131. Kokoshka JM, Fleckenstein AE, Wilkins DG, Hanson GR. Age-dependent differential responses of monoaminergic systems to high doses of methamphetamine. J Neurochem. 2000 Nov;75(5):2095–2102. [PubMed] [Google Scholar]132. Riddle EL, Kokoshka JM, Wilkins DG, Hanson GR, Fleckenstein AE. Tolerance to the neurotoxic effects of methamphetamine in young rats. Eur J Pharmacol. 2002 Jan 25;435(2−3):181–185. [PubMed] [Google Scholar]133. Biederman J, Monuteaux MC, Spencer T, Wilens TE, Macpherson HA, Faraone SV. Stimulant therapy and risk for subsequent substance use disorders in male adults with ADHD: a naturalistic controlled 10-year follow-up study. Am J Psychiatry. 2008 May;165(5):597–603. [PubMed] [Google Scholar]134. Berman SM, O'Neill J, Fears S, Bartzokis G, London ED. Abuse of Amphetamines and Structural Abnormalities in Brain. In: Uhl G, editor. Addiction Reviews. Vol. 1. NY Academy of Sciances; New York: 2008. [PMC free article] [PubMed] [Google Scholar]135. Jaffe JH. Drug addiction and drug abuse. In: Goodman LG, A.G., editors. Pharmacological Basis of Therapeutics. McMillan; New York: 1985. pp. 284–324. [Google Scholar]136. Volkow NDC, L.W., Wang GJ, Fowler JS, Franceschi D, Gatley SJ, Wong CT, Hitzemann R, Pappas NR. In vivo evidence that methamphetamine abuse produces long lasting changes in dopamine transporters in human brain. J Nucl Med, SUPPL. 1999;(40) [Google Scholar]137. McCann UD, Wong DF, Yokoi F, Villemagne V, Dannals RF, Ricaurte GA. Reduced striatal dopamine transporter density in abstinent methamphetamine and methcathinone users: evidence from positron emission tomography studies with [11C]WIN-35,428. J Neurosci. 1998 Oct 15;18(20):8417–8422. [PubMed] [Google Scholar]138. Simon SL, Richardson K, Dacey J, Glynn S, Domier CP, Rawson RA, et al. A comparison of patterns of methamphetamine and cocaine use. J Addict Dis. 2002;21(1):35–44. [PubMed] [Google Scholar]139. Wilson JM, Kalasinsky KS, Levey AI, Bergeron C, Reiber G, Anthony RM, et al. Striatal dopamine nerve terminal markers in human, chronic methamphetamine users. Nat Med. 1996 Jun;2(6):699–703. [PubMed] [Google Scholar]140. Kalechstein AD, Newton TF, Longshore D, Anglin MD, van Gorp WG, Gawin FH. Psychiatric comorbidity of methamphetamine dependence in a forensic sample. J Neuropsychiatry Clin Neurosci. 2000;12(4):480–484. Fall. [PubMed] [Google Scholar]141. Volkow ND, Chang L, Wang GJ, Fowler JS, Leonido-Yee M, Franceschi D, et al. Association of dopamine transporter reduction with psychomotor impairment in methamphetamine abusers. Am J Psychiatry. 2001 Mar;158(3):377–382. [PubMed] [Google Scholar]142. Melega WP, Cho AK, Harvey D, Lacan G. Methamphetamine blood concentrations in human abusers: application to pharmacokinetic modeling. Synapse. 2007 Apr;61(4):216–220. [PubMed] [Google Scholar]143. Chang L, Alicata D, Ernst T, Volkow N. Structural and metabolic brain changes in the striatum associated with methamphetamine abuse. Addiction. 2007;102(Suppl 1):16–32. [PubMed] [Google Scholar]144. Moszczynska A, Fitzmaurice P, Ang L, Kalasinsky KS, Schmunk GA, Peretti FJ, et al. Why is parkinsonism not a feature of human methamphetamine users? Brain. 2004;127:363–370. [PubMed] [Google Scholar]145. Wilson JM, Kalasinsky KS, Levey AI, Bergeron C, Reiber G, Anthony RM, et al. Striatal dopamine nerve terminal markers in human, chronic methamphetamine users. NatMed. 1996;2(6):699–703. [PubMed] [Google Scholar]146. Frey K, Kilbourn M, Robinson T. Reduced striatal vesicular monoamine transporters after neurotoxic but not after behaviorally-sensitizing doses of methamphetamine. European Journal of Pharmacology. 1997;334(2−3):273–279. [PubMed] [Google Scholar]147. Fumagalli F, Gainetdinov RR, Wang YM, Valenzano KJ, Miller GW, Caron MG. Increased methamphetamine neurotoxicity in heterozygous vesicular monoamine transporter 2 knock-out mice. Journal of Neuroscience. 1999;19(7):2424–2431. [PMC free article] [PubMed] [Google Scholar]148. Harvey DC, Lacan G, Tanious SP, Melega WP. Recovery from methamphetamine induced long-term nigrostriatal dopaminergic deficits without substantia nigra cell loss. Brain Research. 2000;871:259–270. [PubMed] [Google Scholar]149. Hogan KA, Staal RG, Sonsalla PK. Analysis of VMAT2 binding after methamphetamine or MPTP treatment: disparity between homogenates and vesicle preparations. Journal of Neurochemistry. 2000;74(5):2217–2220. [PubMed] [Google Scholar]150. Kilbourn MR, Frey KA, Vander BT, Sherman PS. Effects of dopaminergic drug treatments on in vivo radioligand binding to brain vesicular monoamine transporters. NuclMedBiol. 1996;23(4):467–471. [PubMed] [Google Scholar]151. Miller GW, Gainetdinov RR, Levey AI, Caron MG. Dopamine transporters and neuronal injury. Trends in Pharmacological Science. 1999;20(10):424–429. [PubMed] [Google Scholar]152. Vander BT, Kilbourn M, Desmond T, Kuhl D, Frey K. The vesicular monoamine transporter is not regulated by dopaminergic drug treatments. European Journal of Pharmacology. 1995;294(2−3):577–583. [PubMed] [Google Scholar]153. London ED, Simon SL, Berman SM, Mandelkern MA, Lichtman AM, Bramen J, et al. Mood disturbances and regional cerebral metabolic abnormalities in recently abstinent methamphetamine abusers. Archives of General Psychiatry. 2004;61:73–84. [PubMed] [Google Scholar]154. Berman SM, Voytek B, Mandelkern MA, Hassid BD, Isaacson A, Monterosso J, et al. Changes in cerebral glucose metabolism during early abstinence from chronic methamphetamine abuse. Molecular Psychiatry. 2007 Oct 16; [Epub ahead of print] [PMC free article] [PubMed] [Google Scholar]155. Volkow ND, Chang L, Wang GJ, Fowler JS, Franceschi D, Sedler MJ, et al. Higher cortical and lower subcortical metabolism in detoxified methamphetamine abusers. American Journal of Psychiatry. 2001;158(3):383–389. [PubMed] [Google Scholar]156. Wang GJ, Volkow ND, Chang L, Miller E, Sedler M, Hitzemann R, et al. Partial recovery of brain metabolism in methamphetamine abusers after protracted abstinence. American Journal of Psychiatry. 2004;161(2):242–248. [PubMed] [Google Scholar]157. Thompson PM, Hayashi K, Simon SL, Geaga JA, Hong MS, Sui Y, et al. Structural abnormalities in the brains of human subjects who use methamphetamine. J Neurosci. 2004;24(26):6028–6036. [PMC free article] [PubMed] [Google Scholar]158. Chang L, Cloak C, Patterson K, Grob C, Miller EN, Ernst T. Enlarged striatum in abstinent methamphetamine abusers: a possible compensatory response. Biological Psychiatry. 2005;57(9):967–974. [PMC free article] [PubMed] [Google Scholar]159. Jernigan TL, Gamst AC, Archibald SL, Fennema-Notestine C, Mindt MR, Marcotte TD, et al. Effects of methamphetamine dependence and HIV infection on cerebral morphology. Am J Psychiatry. 2005;162(8):1461–1472. [PubMed] [Google Scholar]160. Ernst T, Chang L, Leonido-Yee M, Speck O. Evidence for long-term neurotoxicity associated with methamphetamine abuse: A 1 H MRS study. Neurology. 2000;54(6):1344–1349. [PubMed] [Google Scholar]161. Nordahl TE, Salo R, Natsuaki Y, Galloway GP, Waters C, Moore CD, et al. Methamphetamine users in sustained abstinence: a proton magnetic resonance spectroscopy study. Arch Gen Psychiatry. 2005;62(4):444–452. [PubMed] [Google Scholar]162. Sekine Y, Iyo M, Ouchi Y, Matsunaga T, Tsukada H, Okada H, et al. Methamphetamine-related psychiatric symptoms and reduced brain dopamine transporters studied with PET. Am J Psychiatry. 2001;158(8):1206–1214. [PubMed] [Google Scholar]163. Sekine Y, Minabe Y, Kawai M, Suzuki K, Iyo M, Isoda H, et al. Metabolite alterations in basal ganglia associated with methamphetamine-related psychiatric symptoms: A proton MRS study. Neuropsychopharmacology. 2002;27(3):454–461. [PubMed] [Google Scholar]164. Sekine Y, Minabe Y, Ouchi Y, Takei N, Iyo M, Nakamura K, et al. Association of dopamine transporter loss in the orbitofrontal and dorsolateral prefrontal cortices with methamphetamine-related psychiatric symptoms. American Journal of Psychiatry. 2003;160:1699–1701. [PubMed] [Google Scholar]165. Sekine Y, Ouchi Y, Takei N, Yoshikawa E, Nakamura K, Futatsubashi M, et al. Brain Serotonin Transporter Density and Aggression in Abstinent Methamphetamine Abusers. Arch Gen Psychiatry. 2006;63(1):90–100. [PubMed] [Google Scholar]166. Volkow ND, Chang L, Wang GJ, Fowler JS, Leonido-Yee M, Franceschi D, et al. Association of dopamine transporter reduction with psychomotor impairment in methamphetamine abusers. American Journal of Psychiatry. 2001;158(3):377–382. [PubMed] [Google Scholar]167. Volkow ND, Chang L, Wang GJ, Fowler JS, Ding YS, Sedler M, et al. Low level of brain dopamine D2 receptors in methamphetamine abusers: Association with metabolism in orbitofrontal cortex. American Journal of Psychiatry. 2001;158(12):2015–2021. [PubMed] [Google Scholar]168. London ED, Berman S, Voytek B, Simon SL, Monterosso J, Geaga JA, et al. Cerebral metabolic dysfunction and impaired vigilance in recently abstinent methamphetamine abusers. Biological Psychiatry. 2005;58:770–778. [PubMed] [Google Scholar]169. Hwang J, Lyoo IK, Kim SJ, Sung YH, Bae S, Cho SN, et al. Decreased cerebral blood flow of the right anterior cingulate cortex in long-term and short-term abstinent methamphetamine users. Drug and Alcohol Dependence. 2006;82(2):177–181. [PubMed] [Google Scholar]170. Volkow ND, Chang L, Wang GJ, Fowler JS, Franceschi D, Sedler M, et al. Loss of dopamine transporters in methamphetamine abusers recovers with protracted abstinence. Journal of Neuroscience. 2001;21(23):9414–9418. [PMC free article] [PubMed] [Google Scholar]171. McGregor C, Srisurapanont M, Jittiwutikarn J, Laobhripatr S, Wongtan T, White JM. The nature, time course and severity of methamphetamine withdrawal. Addiction. 2005;100(9):1320–1329. [PubMed] [Google Scholar]172. Newton TF, Kalechstein AD, Duran S, Vansluis N, Ling W. Methamphetamine abstinence syndrome: Preliminary findings. American Journal on Addictions. 2002;13:248–255. [PubMed] [Google Scholar]173. Pennypacker KR, Kassed CA, Eidizadeh S, O'Callaghan JP. Brain injury: prolonged induction of transcription factors. Acta NeurobiolExp(Wars) 2000;60(4):515–530. [PubMed] [Google Scholar]174. Nieuwenhuis S, Aston-Jones G, Cohen JD. Decision making, the P3, and the locus coeruleus-norepinephrine system. Psychological Bulletin. 2005;131(4):510–532. [PubMed] [Google Scholar]175. Polich J, Criado JR. Neuropsychology and neuropharmacology of P3a and P3b. International Journal of Psychophysiology. 2006;60(2):172–185. [PubMed] [Google Scholar]176. Takeuchi S, Jodo E, Suzuki Y, Matsuki T, Niwa S, Kayama Y. Effects of Repeated Administration of Methamphetamine on P3-like Potentials in Rats. International Journal of Psychophysiology. 1999;32(3):183–192. [PubMed] [Google Scholar]177. Volkow ND, Gur RC, Wang GJ, Fowler JS, Moberg PJ, Ding YS, et al. Association between decline in brain dopamine activity with age and cognitive and motor impairment in healthy individuals. American Journal of Psychiatry. 1998;155(3):344–349. [PubMed] [Google Scholar]178. Volkow ND, Logan J, Fowler JS, Wang GJ, Gur RC, Wong C, et al. Association between age-related decline in brain dopamine activity and impairment in frontal and cingulate metabolism. Am J Psychiatry. 2000;157:75–80. [PubMed] [Google Scholar]179. Volkow ND, Fowler JS, Wang GJ, Hitzemann R, Logan J, Schlyer DJ, et al. Decreased dopamine D2 receptor availability is associated with reduced frontal metabolism in cocaine abusers. Synapse. 1993;14:169–177. [PubMed] [Google Scholar]180. Unterharnscheidt F. A neurologist's reflections on boxing. V. Conclude remarks. RevNeurol. 1995;23(123):1027–1032. [PubMed] [Google Scholar]181. Kochunov P, Thompson PM, Coyle TR, Lancaster JL, Kochunov V, Royall D, et al. Relationship among neuroimaging indices of cerebral health during normal aging. HumBrain Mapp. 2008;29(1):36–45. [PubMed] [Google Scholar]182. Castellanos FX, Lee PP, Sharp W, Jeffries NO, Greenstein DK, Clasen LS, et al. Developmental trajectories of brain volume abnormalities in children and adolescents with attention-deficit/hyperactivity disorder. Journal of the American Medical Association. 2002;288:1740–1748. [PubMed] [Google Scholar]183. The MTA Cooperative Group. Multimodal Treatment Study of Children with ADHD A 14-month randomized clinical trial of treatment strategies for attention-deficit/hyperactivity disorder. Archives of General Psychiatry. 1999;56(12):1073–1086. [PubMed] [Google Scholar]184. Kuczenski R, Segal DS. Effects of methylphenidate on extracellular dopamine, serotonin, and norepinephrine: comparison with amphetamine. Journal of Neurochemistry. 1997;68(5):2032–2037. [PubMed] [Google Scholar]185. Schiffer WK, Volkow ND, Fowler JS, Alexoff DL, Logan J, Dewey SL. Therapeutic doses of amphetamine or methylphenidate differentially increase synaptic and extracellular dopamine. Synapse. 2006;59(4):243–251. [PubMed] [Google Scholar]186. Strakowski SM, Sax KW, Rosenberg HL, DelBello MP, Adler CM. Human response to repeated low-dose d-amphetamine: evidence for behavioral enhancement and tolerance. Neuropsychopharmacology. 2001;25(4):548–554. [PubMed] [Google Scholar]187. Becker JB. Gender differences in dopaminergic function in striatum and nucleus accumbens. Pharmacology Biochemistry and Behavior. 1999;64(4):803–812. [PubMed] [Google Scholar]188. Robinson TE, Becker JB. Enduring changes in brain and behavior produced by chronic amphetamine administration: a review and evaluation of animal models of amphetamine psychosis. Brain Res. 1986 Jun;396(2):157–198. [PubMed] [Google Scholar]189. Kuczenski R, Segal DS. Stimulant actions in rodents: implications for attention-deficit/hyperactivity disorder treatment and potential substance abuse. Biol Psychiatry. 2005 Jun 1;57(11):1391–1396. [PubMed] [Google Scholar]190. Angrist B, Gershon S. Dopamine and psychotic states: preliminary remarks. Advances in Biochemical Psychopharmacology. 1974;12(0):211–219. [PubMed] [Google Scholar]191. Curran C, Byrappa N, McBride A. Stimulant psychosis: systematic review. British Journal of Psychiatry. 2004;185:196–204. [PubMed] [Google Scholar]192. Griffith J. A study of illicit amphetamine drug traffic in Oklahoma City. American Journal of Psychiatry. 1966;123(5):560–569. [PubMed] [Google Scholar]193. McKetin R, McLaren J, Lubman DI, Hides L. The prevalence of psychotic symptoms among methamphetamine users. Addiction. 2006;101(10):1473–1478. [PubMed] [Google Scholar]194. Hartel-Petri R, Rodler R, Schmeisser U, Steinmann J, Wolfersdorf M. [Increasing prevalence of amphetamine--and methamphetamine-induced psychosis]. PsychiatrPrax. 2005;32(1):13–17. [PubMed] [Google Scholar]195. Yui K, Ikemoto S, Ishiguro T, Goto K. Studies of amphetamine or methamphetamine psychosis in Japan: relation of methamphetamine psychosis to schizophrenia. Annals of the New York Academy of Sciences. 2000;914:1–12. [PubMed] [Google Scholar]196. Yui K, Ikemoto S, Goto K. Factors for susceptibility to episode recurrence in spontaneous recurrence of methamphetamine psychosis. Annals of the New York Academy of Sciences. 2002;965:292–304. [PubMed] [Google Scholar]197. Ujike H, Harano M, Inada T, Yamada M, Komiyama T, Sekine Y, et al. Nine- or fewer repeat alleles in VNTR polymorphism of the dopamine transporter gene is a strong risk factor for prolonged methamphetamine psychosis. PharmacogenomicsJ. 2003;3(4):242–247. [PubMed] [Google Scholar]198. Ujike H, Japanese Genetics Initiative for Drug Abuse (JGIDA) Nihon Shinkei Seishin Yakurigaku Zasshi. 2004 Oct;24(5):299–302. [PubMed] [Google Scholar]199. Downes MA, Whyte IM. Amphetamine-induced movement disorder. EmergMedAustralas. 2005;17(3):277–280. [PubMed] [Google Scholar]200. Anggard E, Jonsson LE, Hogmark AL, Gunne LM. Amphetamine metabolism in amphetamine psychosis. Clinical Pharmacology and Therapeutics. 1973;14(5):870–880. [PubMed] [Google Scholar]201. Sanga M, Younis IR, Tirumalai PS, Bland TM, Banaszewska M, Konat GW, et al. Epoxidation of the methamphetamine pyrolysis product, trans-phenylpropene, to transphenylpropylene oxide by CYP enzymes and stereoselective glutathione adduct formation. Toxicology and Applied Pharmacology. 2006;211(2):148–156. [PubMed] [Google Scholar]202. Cherland E, Fitzpatrick R. Psychotic side effects of psychostimulants: a 5-year review. CanJPsychiatry. 1999;44(8):811–813. [PubMed] [Google Scholar]203. Masand P, Pickett P, Murray GB. Psychostimulants for secondary depression in medical illness. Psychosomatics. 1991;32(2):203–208. [PubMed] [Google Scholar]204. Murray JB. Psychophysiological aspects of amphetamine-methamphetamine abuse. JPsychol. 1998;132(2):227–237. [PubMed] [Google Scholar]205. Polchert SE, Morse RM. Pemoline abuse. Journal of the American Medical Association. 1985;254(7):946–947. [PubMed] [Google Scholar]206. Surles LK, May HJ, Garry JP. Adderall-induced psychosis in an adolescent. JAmBoardFamPract. 2002;15(6):498–500. [PubMed] [Google Scholar]207. Ross RG. Psychotic and manic-like symptoms during stimulant treatment of attention deficit hyperactivity disorder. American Journal of Psychiatry. 2006;163(7):1149–1152. [PubMed] [Google Scholar]208. Auger RR, Goodman SH, Silber MH, Krahn LE, Pankratz VS, Slocumb NL. Risks of high-dose stimulants in the treatment of disorders of excessive somnolence: a case-control study. Sleep. 2005;28(6):667–672. [PubMed] [Google Scholar]209. Pawluk LK, Hurwitz TD, Schluter JL, Ullevig C, Mahowald MW. Psychiatric morbidity in narcoleptics on chronic high dose methylphenidate therapy. Journal of Nervous and Mental Disease. 1995;183(1):45–48. [PubMed] [Google Scholar]210. Tsuang MT, Lyons MJ, Eisen SA, Goldberg J, True W, Lin N, et al. Genetic influences on DSM-III-R drug abuse and dependence: a study of 3,372 twin pairs. AmJMedGenet. 1996;67(5):473–477. [PubMed] [Google Scholar]211. van den Bree MB, Johnson EO, Neale MC, Pickens RW. Genetic and environmental influences on drug use and abuse/dependence in male and female twins. Drug and Alcohol Dependence. 1998;52(3):231–241. [PubMed] [Google Scholar]212. Suzuki A, Nakamura K, Sekine Y, Minabe Y, Takei N, Suzuki K, et al. An association study between catechol-O-methyl transferase gene polymorphism and methamphetamine psychotic disorder. PsychiatrGenet. 2006;16(4):133–138. [PubMed] [Google Scholar]213. Matsuzawa D, Hashimoto K, Miyatake R, Shirayama Y, Shimizu E, Maeda K, et al. Identification of functional polymorphisms in the promoter region of the human PICK1 gene and their association with methamphetamine psychosis. American Journal of Psychiatry. 2007;164(7):1105–1114. [PubMed] [Google Scholar]214. Madras BK, Marwah J, Teitelbaum H. Advances in Neurodegenerative Disorders. Vol. 1. Prominent Press; Scottsdale, Arizona: 1998. Imaging the dopamine transporter: A window on dopamine neurons. pp. 229–253. [Google Scholar]215. Yatin SM, Miller GM, Norton C, Madras BK. Dopamine transporter-dependent induction of C-Fos in HEK cells. Synapse. 2002;45(1):52–65. [PubMed] [Google Scholar]216. Silverstone PH, Asghar SJ, O'Donnell T, Ulrich M, Hanstock CC. Lithium and valproate protect against dextro-amphetamine induced brain choline concentration changes in bipolar disorder patients. World JBiolPsychiatry. 2004;5(1):38–44. [PubMed] [Google Scholar]217. Wan FJ, Shiah IS, Lin HC, Huang SY, Tung CS. Nomifensine attenuates d-amphetamine-induced dopamine terminal neurotoxicity in the striatum of rats. Chin JPhysiol. 2000;43(2):69–74. [PubMed] [Google Scholar]218. Escubedo E, Chipana C, Perez-Sanchez M, Camarasa J, Pubill D. Methyllycaconitine prevents methamphetamine-induced effects in mouse striatum: involvement of alpha7 nicotinic receptors. JPharmacolExpTher. 2005;315(2):658–667. [PubMed] [Google Scholar]219. Klongpanichapak S, Govitrapong P, Sharma SK, Ebadi M. Attenuation of cocaine and methamphetamine neurotoxicity by coenzyme Q10. Neurochemical Research. 2006;31(3):303–311. [PubMed] [Google Scholar]220. Wu PH, Shen YC, Wang YH, Chi CW, Yen JC. Baicalein attenuates methamphetamine-induced loss of dopamine transporter in mouse striatum. Toxicology. 2006;226(2−3):238–245. [PubMed] [Google Scholar]221. Klongpanichapak S, Phansuwan-Pujito P, Ebadi M, Govitrapong P. Melatonin protects SK-N-SH neuroblastoma cells from amphetamine-induced neurotoxicity. JPineal Res. 2007;43(1):65–73. [PubMed] [Google Scholar]222. Achat-Mendes C, Anderson KL, Itzhak Y. Impairment in consolidation of learned place preference following dopaminergic neurotoxicity in mice is ameliorated by N-acetylcysteine but not D1 and D2 dopamine receptor agonists. Neuropsychopharmacology. 2007;32(3):531–541. [PubMed] [Google Scholar]223. Iversen LL, Oxford University P . Speed, Ecstasy, Ritalin: The Science of Amphetamines. New York, NY: 2006. [Google Scholar]224. Rykhlevskaia E, Gratton G, Fabiani M. Combining structural and functional neuroimaging data for studying brain connectivity: A review. Psychophysiology. 2008;45(2):173–87. [PubMed] [Google Scholar]225. Apostolova LG, Thompson PM. Brain mapping as a tool to study neurodegeneration. Neurotherapeutics. 2007;4(3):387–400. [PMC free article] [PubMed] [Google Scholar]226. Hutton C, De Vita E, Ashburner J, Deichmann R, Turner R. Voxel-based cortical thickness measurements in MRI. Neuroimage. 2008 May 1;40(4):1701–1710. [PMC free article] [PubMed] [Google Scholar]227. de Win MM, Reneman L, Jager G, Vlieger EJ, Olabarriaga SD, Lavini C, et al. A prospective cohort study on sustained effects of low-dose ecstasy use on the brain in new ecstasy users. Neuropsychopharmacology. 2007 Feb;32(2):458–470. [PubMed] [Google Scholar]Formats:Article | PubReader | ePub (beta) | PDF (125K) | CitationShareShare on Facebook Facebook Share on Twitter Twitter Share on Google Plus Google+Save itemsView more optionsSimilar articles in PubMedNTP-CERHR monograph on the potential human reproductive and developmental effects of amphetamines.[NTP CERHR MON. 2005]Psychosis with Methylphenidate or Amphetamine in Patients with ADHD.[N Engl J Med. 2019]Attention-deficit-hyperactivity disorder: an update.[Pharmacotherapy. 2009]Current pharmacotherapy of attention deficit hyperactivity disorder.[Drugs Today (Barc). 2013]Evolution of the treatment of attention-deficit/hyperactivity disorder in children: a review.[Clin Ther. 2008]See reviews...See all...Cited by other articles in PMCD-Amphetamine Exposure Differentially Disrupts Signaling Across Ontogeny in the Zebrafish[Bioelectricity. 2019]Adolescent Δ9-THC exposure and astrocyte-specific genetic vulnerability converge on NF-κB-COX-2 signaling to impair memory in adulthood.[Biological psychiatry. 2018]Dopaminergic Mechanisms Underlying Normal Variation in Trait Anxiety[The Journal of Neuroscience. 2...]A Review of Psychostimulants for Adults With Depression[Federal Practitioner. 2015]Interplay Between Amphetamine and Activity Level in Gene Networks of the Mouse Striatum[Bioinformatics and Biology Ins...]See all...LinksCompoundMedGenPubMedSubstanceTaxonomyRecent ActivityClearTurn OffPotential Adverse Effects of Amphetamine Treatment on Brain and Behavior: A Revi...Potential Adverse Effects of Amphetamine Treatment on Brain and Behavior: A ReviewNIHPA Author Manuscripts. 2009 Feb; 14(2)123Cannabidiol in Anxiety and Sleep: A Large Case SeriesCannabidiol in Anxiety and Sleep: A Large Case SeriesThe Permanente Journal. 2019; 23()Cannabidiol in Anxiety and Sleep: A Large Case Series.Cannabidiol in Anxiety and Sleep: A Large Case Series.Perm J. 2019;23:18-041. doi: 10.7812/TPP/18-041.PubMedNovel psychoactive substances: overdose of 3-fluorophenmetrazine (3-FPM) and eti...Novel psychoactive substances: overdose of 3-fluorophenmetrazine (3-FPM) and etizolam in a 33-year-old man.BMJ Case Rep. 2018 Jun 8;2018. pii: bcr-2018-224995. doi: 10.1136/bcr-2018-224995.PubMedRapid and sustained symptom reduction following psilocybin treatment for anxiety...Rapid and sustained symptom reduction following psilocybin treatment for anxiety and depression in patients with life-threatening cancer: a randomized controlled trialSAGE Choice. 2016 Dec; 30(12)1165See more...Review New insights into the mechanism of action of amphetamines.[Annu Rev Pharmacol Toxicol. 2007]Review The dopamine transporter and attention-deficit/hyperactivity disorder.[Biol Psychiatry. 2005]Review What keeps us awake: the neuropharmacology of stimulants and wakefulness-promoting medications.[Sleep. 2004]Review New insights into the mechanism of action of amphetamines.[Annu Rev Pharmacol Toxicol. 2007]Stimulant drug treatment of hyperactivity: biochemical correlates.[Clin Pharmacol Ther. 1990]Hippocampus norepinephrine, caudate dopamine and serotonin, and behavioral responses to the stereoisomers of amphetamine and methamphetamine.[J Neurosci. 1995]Cerebral glucose metabolism in adults with hyperactivity of childhood onset.[N Engl J Med. 1990]Review Diagnosis and treatment of attention-deficit/hyperactivity disorder in children and adolescents. Council on Scientific Affairs, American Medical Association.[JAMA. 1998]Pharmacokinetics of SLI381 (ADDERALL XR), an extended-release formulation of Adderall.[J Am Acad Child Adolesc Psychiatry. 2003]Lisdexamfetamine dimesylate: the first prodrug stimulant.[Psychiatry (Edgmont). 2007]Review New formulations of stimulants for attention-deficit hyperactivity disorder: therapeutic potential.[CNS Drugs. 2004]Attention-deficit/hyperactivity disorder in adults.[JAMA. 2004]What are the long-term effects of methylphenidate treatment?[Biol Psychiatry. 2003]Stimulant drug treatment of hyperactivity: biochemical correlates.[Clin Pharmacol Ther. 1990]Review Narcolepsy and its treatment with stimulants. ASDA standards of practice.[Sleep. 1994]Relative efficacy of drugs for the treatment of sleepiness in narcolepsy.[Sleep. 1991]Review NTP-CERHR monograph on the potential human reproductive and developmental effects of amphetamines.[NTP CERHR MON. 2005]Clinical practice guideline: treatment of the school-aged child with attention-deficit/hyperactivity disorder.[Pediatrics. 2001]Side effects of methylphenidate and dexamphetamine in children with attention deficit hyperactivity disorder: a double-blind, crossover trial.[Pediatrics. 1997]Pharmacokinetics of SLI381 (ADDERALL XR), an extended-release formulation of Adderall.[J Am Acad Child Adolesc Psychiatry. 2003]Review Cardiovascular manifestations of substance abuse: part 2: alcohol, amphetamines, heroin, cannabis, and caffeine.[Heart Dis. 2003]Review Concomitant psychotropic medication for youths.[Am J Psychiatry. 2003]Multiple psychotropic medication use for youths: a two-state comparison.[J Child Adolesc Psychopharmacol. 2005]Review Diagnosis and treatment of attention-deficit/hyperactivity disorder in children and adolescents. Council on Scientific Affairs, American Medical Association.[JAMA. 1998]Review Current concepts on the neurobiology of Attention-Deficit/Hyperactivity Disorder.[J Atten Disord. 2002]Comparing the efficacy of medications for ADHD using meta-analysis.[MedGenMed. 2006]Parents report on stimulant-treated children in the Netherlands: initiation of treatment and follow-up care.[J Child Adolesc Psychopharmacol. 2006]Patterns of psychostimulant prescribing to children with ADHD in Western Australia: variations in age, gender, medication type and dose prescribed.[Aust N Z J Public Health. 2007]Patterns of psychostimulant prescribing to children with ADHD in Western Australia: variations in age, gender, medication type and dose prescribed.[Aust N Z J Public Health. 2007]Diagnosis and treatment of adult attention-deficit/hyperactivity disorder at US ambulatory care visits from 1996 to 2003.[Curr Med Res Opin. 2006]Health care costs of adults treated for attention-deficit/hyperactivity disorder who received alternative drug therapies.[J Manag Care Pharm. 2007]Review New formulations of stimulants for attention-deficit hyperactivity disorder: therapeutic potential.[CNS Drugs. 2004]Attention-deficit/hyperactivity disorder in adults.[JAMA. 2004]Plasma levels of d-amphetamine in hyperactive children. Serial behavior and motor responses.[Psychopharmacology (Berl). 1979]Review Therapeutic advances in narcolepsy.[Sleep Med. 2007]Review Drugs of abuse and the aging brain.[Neuropsychopharmacology. 2008]Substance abuse treatment need among older adults in 2020: the impact of the aging baby-boom cohort.[Drug Alcohol Depend. 2003]Review Current research on methamphetamine-induced neurotoxicity: animal models of monoamine disruption.[J Pharmacol Sci. 2003]Amphetamine sensitization alters dendritic morphology in prefrontal cortical pyramidal neurons in the non-human primate.[Neuropsychopharmacology. 2007]Review Interactions between methamphetamine and environmental stress: role of oxidative stress, glutamate and mitochondrial dysfunction.[Addiction. 2007]Escalating dose-binge treatment with methylphenidate: role of serotonin in the emergent behavioral profile.[J Pharmacol Exp Ther. 1999]Methylphenidate and brain dopamine neurotoxicity.[Brain Res. 1997]Hippocampus norepinephrine, caudate dopamine and serotonin, and behavioral responses to the stereoisomers of amphetamine and methamphetamine.[J Neurosci. 1995]Chemical release of dopamine from striatal homogenates: evidence for an exchange diffusion model.[J Pharmacol Exp Ther. 1979]Repeated binge exposures to amphetamine and methamphetamine: behavioral and neurochemical characterization.[J Pharmacol Exp Ther. 1997]Recovery of striatal dopamine function after acute amphetamine- and methamphetamine-induced neurotoxicity in the vervet monkey.[Brain Res. 1997]Early methylphenidate administration to young rats causes a persistent reduction in the density of striatal dopamine transporters.[J Child Adolesc Psychopharmacol. 2001]Repeated binge exposures to amphetamine and methamphetamine: behavioral and neurochemical characterization.[J Pharmacol Exp Ther. 1997]Amphetamine treatment similar to that used in the treatment of adult attention-deficit/hyperactivity disorder damages dopaminergic nerve endings in the striatum of adult nonhuman primates.[J Pharmacol Exp Ther. 2005]Differential effects of methylphenidate and dextroamphetamine on the motor activity level of hyperactive children.[Neuropsychopharmacology. 1989]Pharmacokinetics of SLI381 (ADDERALL XR), an extended-release formulation of Adderall.[J Am Acad Child Adolesc Psychiatry. 2003]Amphetamine treatment similar to that used in the treatment of adult attention-deficit/hyperactivity disorder damages dopaminergic nerve endings in the striatum of adult nonhuman primates.[J Pharmacol Exp Ther. 2005]Pharmacokinetics of SLI381 (ADDERALL XR), an extended-release formulation of Adderall.[J Am Acad Child Adolesc Psychiatry. 2003]A pharmacokinetic/pharmacodynamic study comparing a single morning dose of adderall to twice-daily dosing in children with ADHD.[J Am Acad Child Adolesc Psychiatry. 2003]Repeated binge exposures to amphetamine and methamphetamine: behavioral and neurochemical characterization.[J Pharmacol Exp Ther. 1997]Review Interactions between methamphetamine and environmental stress: role of oxidative stress, glutamate and mitochondrial dysfunction.[Addiction. 2007]Effects of neonatal and prepubertal hormonal manipulations upon estrogen neuroprotection of the nigrostriatal dopaminergic system within female and male mice.[Neuroscience. 2005]Effects of a cold environment or age on methamphetamine-induced dopamine release in the caudate putamen of female rats.[Pharmacol Biochem Behav. 1993]Age-related toxicity in prefrontal cortex and caudate-putamen complex of gerbils (Meriones unguiculatus) after a single dose of methamphetamine.[Neuropharmacology. 1991]3H-amphetamine concentrations in the brains of young and aged rats: implications for assessment of drug effects in aged animals.[Neurobiol Aging. 1980]Escalating dose pretreatment induces pharmacodynamic and not pharmacokinetic tolerance to a subsequent high-dose methamphetamine binge.[Synapse. 2006]Escalating dose methamphetamine pretreatment alters the behavioral and neurochemical profiles associated with exposure to a high-dose methamphetamine binge.[Neuropsychopharmacology. 2003]Escalating dose pretreatment induces pharmacodynamic and not pharmacokinetic tolerance to a subsequent high-dose methamphetamine binge.[Synapse. 2006]Influence of age and time interval between death and autopsy on dopamine and 3-methoxytyramine levels in human basal ganglia.[J Neural Transm. 1976]Review Update on amphetamine neurotoxicity and its relevance to the treatment of ADHD.[J Atten Disord. 2007]Repeated binge exposures to amphetamine and methamphetamine: behavioral and neurochemical characterization.[J Pharmacol Exp Ther. 1997]Recovery of striatal dopamine function after acute amphetamine- and methamphetamine-induced neurotoxicity in the vervet monkey.[Brain Res. 1997]Review NTP-CERHR monograph on the potential human reproductive and developmental effects of amphetamines.[NTP CERHR MON. 2005]Review Growth on stimulant medication; clarifying the confusion: a review.[Arch Dis Child. 2005]Comparative effects of methylphenidate and mixed salts amphetamine on height and weight in children with attention-deficit/hyperactivity disorder.[J Am Acad Child Adolesc Psychiatry. 2006]Does prolonged therapy with a long-acting stimulant suppress growth in children with ADHD?[J Am Acad Child Adolesc Psychiatry. 2006]Effects of stimulant medication on growth rates across 3 years in the MTA follow-up.[J Am Acad Child Adolesc Psychiatry. 2007]Comparative effects of methylphenidate and mixed salts amphetamine on height and weight in children with attention-deficit/hyperactivity disorder.[J Am Acad Child Adolesc Psychiatry. 2006]Stimulant treatment over 5 years: effects on growth.[J Am Acad Child Adolesc Psychiatry. 2006]Effects of stimulant medication on growth rates across 3 years in the MTA follow-up.[J Am Acad Child Adolesc Psychiatry. 2007]Does prolonged therapy with a long-acting stimulant suppress growth in children with ADHD?[J Am Acad Child Adolesc Psychiatry. 2006]Stimulant-related reductions of growth rates in the PATS.[J Am Acad Child Adolesc Psychiatry. 2006]Review NTP-CERHR monograph on the potential human reproductive and developmental effects of amphetamines.[NTP CERHR MON. 2005]Review Treatment for amphetamine dependence and abuse.[Cochrane Database Syst Rev. 2001]Review Biological treatments for amfetamine dependence : recent progress.[CNS Drugs. 2007]Use of d-amphetamine and related central nervous system stimulants in children.[Pediatrics. 1973]Why do we need an Addiction supplement focused on methamphetamine?[Addiction. 2007]Review NTP-CERHR monograph on the potential human reproductive and developmental effects of amphetamines.[NTP CERHR MON. 2005]Analysis of pyrolysis products of methamphetamine.[J Anal Toxicol. 2004]Stimulant use and the potential for abuse in Wisconsin as reported by school administrators and longitudinally followed children.[J Dev Behav Pediatr. 1998]The use, misuse and diversion of prescription stimulants among middle and high school students.[Subst Use Misuse. 2004]Medical use, illicit use and diversion of prescription stimulant medication.[J Psychoactive Drugs. 2006]Medical use, illicit use, and diversion of abusable prescription drugs.[J Am Coll Health. 2006]Medical and nonmedical stimulant use among adolescents: from sanctioned to unsanctioned use.[CMAJ. 2001]Issues of medication administration and control in Iowa schools.[J Sch Health. 2003]Parents report on stimulant-treated children in the Netherlands: initiation of treatment and follow-up care.[J Child Adolesc Psychopharmacol. 2006]Review Therapeutic advances in narcolepsy.[Sleep Med. 2007]Review Narcolepsy.[Neurol Clin. 1996]Reinforcing, subject-rated, performance and physiological effects of methylphenidate and d-amphetamine in stimulant abusing humans.[J Psychopharmacol. 2004]Review Comparing the abuse potential of methylphenidate versus other stimulants: a review of available evidence and relevance to the ADHD patient.[J Clin Psychiatry. 2003]Discriminative stimulus and self-reported effects of methylphenidate, d-amphetamine, and triazolam in methylphenidate-trained humans.[Exp Clin Psychopharmacol. 2005]PET study examining pharmacokinetics, detection and likeability, and dopamine transporter receptor occupancy of short- and long-acting oral methylphenidate.[Am J Psychiatry. 2006]Review When ADHD and substance use disorders intersect: relationship and treatment implications.[Curr Psychiatry Rep. 2007]Do individuals with ADHD self-medicate with cigarettes and substances of abuse? Results from a controlled family study of ADHD.[Am J Addict. 2007]Prospective study of tobacco smoking and substance dependencies among samples of ADHD and non-ADHD participants.[J Learn Disabil. 1998]Review Long-term effects of stimulant medications on the brain: possible relevance to the treatment of attention deficit hyperactivity disorder.[J Child Adolesc Psychopharmacol. 2001]Review When ADHD and substance use disorders intersect: relationship and treatment implications.[Curr Psychiatry Rep. 2007]Does stimulant therapy of attention-deficit/hyperactivity disorder beget later substance abuse? A meta-analytic review of the literature.[Pediatrics. 2003]Medical use, illicit use and diversion of prescription stimulant medication.[J Psychoactive Drugs. 2006]Exposure of adolescent rats to oral methylphenidate: preferential effects on extracellular norepinephrine and absence of sensitization and cross-sensitization to methamphetamine.[J Neurosci. 2002]Early developmental exposure to methylphenidate reduces cocaine-induced potentiation of brain stimulation reward in rats.[Biol Psychiatry. 2005]Enhanced reactivity and vulnerability to cocaine following methylphenidate treatment in adolescent rats.[Neuropsychopharmacology. 2001]Age of methylphenidate treatment initiation in children with ADHD and later substance abuse: prospective follow-up into adulthood.[Am J Psychiatry. 2008]Review The adolescent brain and age-related behavioral manifestations.[Neurosci Biobehav Rev. 2000]Amygdala circuitry in attentional and representational processes.[Trends Cogn Sci. 1999]The locomotor effects of MK801 in the nucleus accumbens of developing and adult rats.[Eur J Pharmacol. 1999]Development of the dopaminergic innervation in the prefrontal cortex of the rat.[J Comp Neurol. 1988]Postnatal development of D1 dopamine receptors in the medial prefrontal cortex, striatum and nucleus accumbens of normal and neonatal 6-hydroxydopamine treated rats: a quantitative autoradiographic analysis.[Brain Res Dev Brain Res. 1991]Review The development of the rat prefrontal cortex. Its size and development of connections with thalamus, spinal cord and other cortical areas.[Prog Brain Res. 1990]Effects of neural stimuli on paraventricular nucleus neurones.[Brain Res Bull. 1985]Subsensitivity to dopaminergic drugs in periadolescent rats: a behavioral and neurochemical analysis.[Brain Res Dev Brain Res. 1998]Augmentation of prefrontal cortical monoaminergic activity inhibits dopamine release in the caudate nucleus: an in vivo neurochemical assessment in the rhesus monkey.[Neuroscience. 1995]Plasma and brain methamphetamine concentrations in neonatal rats.[Neurotoxicol Teratol. 2001]Neurochemical consequences following administration of CNS stimulants to the neonatal rat.[Pharmacol Biochem Behav. 1981]Review Critical periods of vulnerability for the developing nervous system: evidence from humans and animal models.[Environ Health Perspect. 2000]Developmental dissociation of methamphetamine-induced depletion of dopaminergic terminals and astrocyte reaction in rat striatum.[Brain Res Dev Brain Res. 1993]Age-dependent differential responses of monoaminergic systems to high doses of methamphetamine.[J Neurochem. 2000]Tolerance to the neurotoxic effects of methamphetamine in young rats.[Eur J Pharmacol. 2002]Stimulant therapy and risk for subsequent substance use disorders in male adults with ADHD: a naturalistic controlled 10-year follow-up study.[Am J Psychiatry. 2008]Age of methylphenidate treatment initiation in children with ADHD and later substance abuse: prospective follow-up into adulthood.[Am J Psychiatry. 2008]Review Abuse of amphetamines and structural abnormalities in the brain.[Ann N Y Acad Sci. 2008]Psychiatric comorbidity of methamphetamine dependence in a forensic sample.[J Neuropsychiatry Clin Neurosci. 2000]Association of dopamine transporter reduction with psychomotor impairment in methamphetamine abusers.[Am J Psychiatry. 2001]Methamphetamine blood concentrations in human abusers: application to pharmacokinetic modeling.[Synapse. 2007]Review Structural and metabolic brain changes in the striatum associated with methamphetamine abuse.[Addiction. 2007]Review Current research on methamphetamine-induced neurotoxicity: animal models of monoamine disruption.[J Pharmacol Sci. 2003]Why is parkinsonism not a feature of human methamphetamine users?[Brain. 2004]Striatal dopamine nerve terminal markers in human, chronic methamphetamine users.[Nat Med. 1996]Reduced striatal vesicular monoamine transporters after neurotoxic but not after behaviorally-sensitizing doses of methamphetamine.[Eur J Pharmacol. 1997]Analysis of VMAT2 binding after methamphetamine or MPTP treatment: disparity between homogenates and vesicle preparations.[J Neurochem. 2000]Effects of dopaminergic drug treatments on in vivo radioligand binding to brain vesicular monoamine transporters.[Nucl Med Biol. 1996]The vesicular monoamine transporter is not regulated by dopaminergic drug treatments.[Eur J Pharmacol. 1995]Review Structural and metabolic brain changes in the striatum associated with methamphetamine abuse.[Addiction. 2007]Mood disturbances and regional cerebral metabolic abnormalities in recently abstinent methamphetamine abusers.[Arch Gen Psychiatry. 2004]Changes in cerebral glucose metabolism during early abstinence from chronic methamphetamine abuse.[Mol Psychiatry. 2008]Higher cortical and lower subcortical metabolism in detoxified methamphetamine abusers.[Am J Psychiatry. 2001]Partial recovery of brain metabolism in methamphetamine abusers after protracted abstinence.[Am J Psychiatry. 2004]Structural abnormalities in the brains of human subjects who use methamphetamine.[J Neurosci. 2004]Enlarged striatum in abstinent methamphetamine abusers: a possible compensatory response.[Biol Psychiatry. 2005]Effects of methamphetamine dependence and HIV infection on cerebral morphology.[Am J Psychiatry. 2005]Review Abuse of amphetamines and structural abnormalities in the brain.[Ann N Y Acad Sci. 2008]Evidence for long-term neurotoxicity associated with methamphetamine abuse: A 1H MRS study.[Neurology. 2000]Association of dopamine transporter reduction with psychomotor impairment in methamphetamine abusers.[Am J Psychiatry. 2001]Low level of brain dopamine D2 receptors in methamphetamine abusers: association with metabolism in the orbitofrontal cortex.[Am J Psychiatry. 2001]Changes in cerebral glucose metabolism during early abstinence from chronic methamphetamine abuse.[Mol Psychiatry. 2008]Higher cortical and lower subcortical metabolism in detoxified methamphetamine abusers.[Am J Psychiatry. 2001]Mood disturbances and regional cerebral metabolic abnormalities in recently abstinent methamphetamine abusers.[Arch Gen Psychiatry. 2004]Cerebral metabolic dysfunction and impaired vigilance in recently abstinent methamphetamine abusers.[Biol Psychiatry. 2005]Enlarged striatum in abstinent methamphetamine abusers: a possible compensatory response.[Biol Psychiatry. 2005]Decreased cerebral blood flow of the right anterior cingulate cortex in long-term and short-term abstinent methamphetamine users.[Drug Alcohol Depend. 2006]Loss of dopamine transporters in methamphetamine abusers recovers with protracted abstinence.[J Neurosci. 2001]Partial recovery of brain metabolism in methamphetamine abusers after protracted abstinence.[Am J Psychiatry. 2004]Changes in cerebral glucose metabolism during early abstinence from chronic methamphetamine abuse.[Mol Psychiatry. 2008]The nature, time course and severity of methamphetamine withdrawal.[Addiction. 2005]Methamphetamine abstinence syndrome: preliminary findings.[Am J Addict. 2004]Review Brain injury: prolonged induction of transcription factors.[Acta Neurobiol Exp (Wars). 2000]Review Decision making, the P3, and the locus coeruleus-norepinephrine system.[Psychol Bull. 2005]Review Neuropsychology and neuropharmacology of P3a and P3b.[Int J Psychophysiol. 2006]Effects of repeated administration of methamphetamine on P3-like potentials in rats.[Int J Psychophysiol. 1999]Review NTP-CERHR monograph on the potential human reproductive and developmental effects of amphetamines.[NTP CERHR MON. 2005]Amphetamine treatment similar to that used in the treatment of adult attention-deficit/hyperactivity disorder damages dopaminergic nerve endings in the striatum of adult nonhuman primates.[J Pharmacol Exp Ther. 2005]Review Drugs of abuse and the aging brain.[Neuropsychopharmacology. 2008]Association between decline in brain dopamine activity with age and cognitive and motor impairment in healthy individuals.[Am J Psychiatry. 1998]Association between age-related decline in brain dopamine activity and impairment in frontal and cingulate metabolism.[Am J Psychiatry. 2000]Decreased dopamine D2 receptor availability is associated with reduced frontal metabolism in cocaine abusers.[Synapse. 1993]Review A neurologist's reflections on boxing. V. Conclude remarks.[Rev Neurol. 1995]Why is parkinsonism not a feature of human methamphetamine users?[Brain. 2004]Relationship among neuroimaging indices of cerebral health during normal aging.[Hum Brain Mapp. 2008]Developmental trajectories of brain volume abnormalities in children and adolescents with attention-deficit/hyperactivity disorder.[JAMA. 2002]A 14-month randomized clinical trial of treatment strategies for attention-deficit/hyperactivity disorder. The MTA Cooperative Group. Multimodal Treatment Study of Children with ADHD.[Arch Gen Psychiatry. 1999]Side effects of methylphenidate and dexamphetamine in children with attention deficit hyperactivity disorder: a double-blind, crossover trial.[Pediatrics. 1997]Effects of methylphenidate on extracellular dopamine, serotonin, and norepinephrine: comparison with amphetamine.[J Neurochem. 1997]Therapeutic doses of amphetamine or methylphenidate differentially increase synaptic and extracellular dopamine.[Synapse. 2006]Human response to repeated low-dose d-amphetamine: evidence for behavioral enhancement and tolerance.[Neuropsychopharmacology. 2001]Review Gender differences in dopaminergic function in striatum and nucleus accumbens.[Pharmacol Biochem Behav. 1999]Review Enduring changes in brain and behavior produced by chronic amphetamine administration: a review and evaluation of animal models of amphetamine psychosis.[Brain Res. 1986]Review Stimulant actions in rodents: implications for attention-deficit/hyperactivity disorder treatment and potential substance abuse.[Biol Psychiatry. 2005]Diagnosis and treatment of adult attention-deficit/hyperactivity disorder at US ambulatory care visits from 1996 to 2003.[Curr Med Res Opin. 2006]Review Dopamine and psychotic states: preliminary remarks.[Adv Biochem Psychopharmacol. 1974]Review Stimulant psychosis: systematic review.[Br J Psychiatry. 2004]A study of illicit amphetamine drug traffic in Oklahoma City.[Am J Psychiatry. 1966]The prevalence of psychotic symptoms among methamphetamine users.[Addiction. 2006][Increasing prevalence of amphetamine--and methamphetamine-induced psychosis].[Psychiatr Prax. 2005]Review Enduring changes in brain and behavior produced by chronic amphetamine administration: a review and evaluation of animal models of amphetamine psychosis.[Brain Res. 1986]Review Studies of amphetamine or methamphetamine psychosis in Japan: relation of methamphetamine psychosis to schizophrenia.[Ann N Y Acad Sci. 2000]Factors for susceptibility to episode recurrence in spontaneous recurrence of methamphetamine psychosis.[Ann N Y Acad Sci. 2002]Nine- or fewer repeat alleles in VNTR polymorphism of the dopamine transporter gene is a strong risk factor for prolonged methamphetamine psychosis.[Pharmacogenomics J. 2003]Review [Japanese Genetics Initiative for Drug Abuse (JGIDA)].[Nihon Shinkei Seishin Yakurigaku Zasshi. 2004]Review Stimulant psychosis: systematic review.[Br J Psychiatry. 2004]Review Amphetamine-induced movement disorder.[Emerg Med Australas. 2005]Review Abuse of amphetamines and structural abnormalities in the brain.[Ann N Y Acad Sci. 2008]Review NTP-CERHR monograph on the potential human reproductive and developmental effects of amphetamines.[NTP CERHR MON. 2005]Amphetamine metabolism in amphetamine psychosis.[Clin Pharmacol Ther. 1973]Epoxidation of the methamphetamine pyrolysis product, trans-phenylpropene, to trans-phenylpropylene oxide by CYP enzymes and stereoselective glutathione adduct formation.[Toxicol Appl Pharmacol. 2006]Psychotic side effects of psychostimulants: a 5-year review.[Can J Psychiatry. 1999]Adderall-induced psychosis in an adolescent.[J Am Board Fam Pract. 2002]Psychotic and manic-like symptoms during stimulant treatment of attention deficit hyperactivity disorder.[Am J Psychiatry. 2006]Nine- or fewer repeat alleles in VNTR polymorphism of the dopamine transporter gene is a strong risk factor for prolonged methamphetamine psychosis.[Pharmacogenomics J. 2003]Review [Japanese Genetics Initiative for Drug Abuse (JGIDA)].[Nihon Shinkei Seishin Yakurigaku Zasshi. 2004]Risks of high-dose stimulants in the treatment of disorders of excessive somnolence: a case-control study.[Sleep. 2005]Psychiatric morbidity in narcoleptics on chronic high dose methylphenidate therapy.[J Nerv Ment Dis. 1995]Genetic influences on DSM-III-R drug abuse and dependence: a study of 3,372 twin pairs.[Am J Med Genet. 1996]Genetic and environmental influences on drug use and abuse/dependence in male and female twins.[Drug Alcohol Depend. 1998]Review [Japanese Genetics Initiative for Drug Abuse (JGIDA)].[Nihon Shinkei Seishin Yakurigaku Zasshi. 2004]Nine- or fewer repeat alleles in VNTR polymorphism of the dopamine transporter gene is a strong risk factor for prolonged methamphetamine psychosis.[Pharmacogenomics J. 2003]An association study between catechol-O-methyl transferase gene polymorphism and methamphetamine psychotic disorder.[Psychiatr Genet. 2006]Identification of functional polymorphisms in the promoter region of the human PICK1 gene and their association with methamphetamine psychosis.[Am J Psychiatry. 2007]Dopamine transporter-dependent induction of C-Fos in HEK cells.[Synapse. 2002]Review [Japanese Genetics Initiative for Drug Abuse (JGIDA)].[Nihon Shinkei Seishin Yakurigaku Zasshi. 2004]Review Lithium and valproate protect against dextro-amphetamine induced brain choline concentration changes in bipolar disorder patients.[World J Biol Psychiatry. 2004]Nomifensine attenuates d-amphetamine-induced dopamine terminal neurotoxicity in the striatum of rats.[Chin J Physiol. 2000]Methyllycaconitine prevents methamphetamine-induced effects in mouse striatum: involvement of alpha7 nicotinic receptors.[J Pharmacol Exp Ther. 2005]Attenuation of cocaine and methamphetamine neurotoxicity by coenzyme Q10.[Neurochem Res. 2006]Baicalein attenuates methamphetamine-induced loss of dopamine transporter in mouse striatum.[Toxicology. 2006]Melatonin protects SK-N-SH neuroblastoma cells from amphetamine-induced neurotoxicity.[J Pineal Res. 2007]Impairment in consolidation of learned place preference following dopaminergic neurotoxicity in mice is ameliorated by N-acetylcysteine but not D1 and D2 dopamine receptor agonists.[Neuropsychopharmacology. 2007]Review Combining structural and functional neuroimaging data for studying brain connectivity: a review.[Psychophysiology. 2008]Review Brain mapping as a tool to study neurodegeneration.[Neurotherapeutics. 2007]Voxel-based cortical thickness measurements in MRI.[Neuroimage. 2008]A prospective cohort study on sustained effects of low-dose ecstasy use on the brain in new ecstasy users.Neuropsychopharmacology.

Comments from Our Customers

I like the easy of use dashboard options, document preparation and easy to use drag and drop field population.

Justin Miller