Elsevier

Drug and Alcohol Dependence

Volume 152, 1 July 2015, Pages 47-56
Drug and Alcohol Dependence

Full length article
Brain and cognition abnormalities in long-term anabolic-androgenic steroid users

https://doi.org/10.1016/j.drugalcdep.2015.04.023Get rights and content

Highlights

  • Anabolic-androgenic steroids (AAS) cause psychiatric and cognitive abnormalities.

  • We conducted the first systematic brain imaging study of human long-term AAS users.

  • AAS users had larger right amygdalas and reduced right amygdala fMRI connectivity.

  • AAS users also had dorsal anterior cingulate cortex neurochemical abnormalities.

  • AAS use causes brain changes that may underlie psychiatric and cognitive changes.

Abstract

Background

Anabolic-androgenic steroid (AAS) use is associated with psychiatric symptoms including increased aggression as well as with cognitive dysfunction. The brain effects of long-term AAS use have not been assessed in humans.

Methods

This multimodal magnetic resonance imaging study of the brain compared 10 male weightlifters reporting long-term AAS use with 10 age-matched weightlifters reporting no AAS exposure. Participants were administered visuospatial memory tests and underwent neuroimaging. Brain volumetric analyses were performed; resting-state fMRI functional connectivity (rsFC) was evaluated using a region-of-interest analysis focused on the amygdala; and dorsal anterior cingulate cortex (dACC) metabolites were quantified by proton magnetic resonance spectroscopy (MRS).

Results

AAS users had larger right amygdala volumes than nonusers (P = 0.002) and reduced rsFC between right amygdala and frontal, striatal, limbic, hippocampal, and visual cortical areas. Left amygdala volumes were slightly larger in AAS users (P = 0.061) but few group differences were detected in left amygdala rsFC. AAS users also had lower dACC scyllo-inositol levels (P = 0.004) and higher glutamine/glutamate ratios (P = 0.028), possibly reflecting increased glutamate turnover. On a visuospatial cognitive task, AAS users performed more poorly than nonusers, with the difference approaching significance (P = 0.053).

Conclusions

Long-term AAS use is associated with right amygdala enlargement and reduced right amygdala rsFC with brain areas involved in cognitive control and spatial memory, which could contribute to the psychiatric effects and cognitive dysfunction associated with AAS use. The MRS abnormalities we detected could reflect enhanced glutamate turnover and increased vulnerability to neurotoxic or neurodegenerative processes, which could contribute to AAS-associated cognitive dysfunction.

Introduction

Illicit anabolic-androgenic steroids (AAS) use poses a growing public health problem worldwide, with 2.9–4.0 million individuals in the US alone having used these drugs (Pope et al., 2014). Virtually all of these users are male (Kanayama et al., 2007). Although used by elite athletes since the 1950s, AAS did not spread widely to the general population until the 1980s (Kanayama et al., 2008). Thus, even the oldest AAS users, who initiated AAS use as youths in the 1980s, are mostly under age 50 today. As the leading edge of this AAS-user population passes through middle age, adverse general health effects of long-term AAS exposure are becoming increasingly apparent (Pope et al., 2014).

AAS are known to cause acute psychiatric effects such as aggression (Copeland et al., 2000, Perry et al., 2003, Pope et al., 2014), violence, including increased partner violence (Beaver et al., 2008, Choi and Pope, 1994, Middleman et al., 1995, Pope and Katz, 1990, Skarberg et al., 2010, Thiblin and Parlklo, 2002), and impulsive behaviors including risky sexual and other behaviors (Hildebrandt et al., 2014, Middleman et al., 1995, Midgley et al., 2000). We (Kouri et al., 1995, Pope et al., 2000) and others (Su et al., 1993, Yates et al., 1999) have documented such effects in controlled human studies. AAS also increase aggressive behaviors in adolescent and adult rodents (Kalinine et al., 2014, Melloni and Ferris, 1996), which may be associated with reduced glutamate uptake and increased N-methyl-d-aspartate (NMDA) receptor activity (Kalinine et al., 2014).

AAS also may cause chronic cognitive effects. Recently, we reported (Kanayama et al., 2013) that long-term AAS users exhibited deficits on two tests of visuospatial memory from the widely used CANTAB battery (Cambridge Cognition, 2015), and the severity of these deficits was associated with lifetime dose of AAS used. One of these tests, Paired Associates Learning, has previously been shown to predict the development of dementia (Swainson et al., 2001). Consistent with our human findings, rodent studies have shown that AAS exposure can impair performance on the Morris water maze test of spatial learning and memory (Magnusson et al., 2009, Novaes Gomes et al., 2014, Pieretti et al., 2013, Tanehkar et al., 2013). Impaired inhibitory control and attention also were recently reported in men actively taking AAS, with greater impairment found in adolescent-than adult-onset AAS users (Hildebrandt et al., 2014).

While the human brain substrates for these AAS effects have yet to be elucidated, the findings reviewed above suggest that attentional regions of the brain associated with threat reactivity and regulation, such as the amygdala, the hippocampus, and the dorsal anterior cingulate cortex (dACC), may be particularly vulnerable to chronic AAS use. The amygdala is involved in threat processing and aggression (Siever, 2008). The rat amygdala is androgen-sensitive (Cooke et al., 1999, Lynch and Story, 2000) and androgen administration to male rats induces amygdala neurogenesis and neuronal soma and astrocyte volume and complexity increases (Fowler et al., 2003, Cooke et al., 1999, Johnson et al., 2008, Johnson et al., 2012). Functional MRI (fMRI) studies in healthy men report positive associations between amygdala reactivity to angry or fearful faces and levels of the endogenous AAS testosterone (Derntl et al., 2009). Similarly, testosterone administration to healthy men acutely increased amygdala reactivity to angry faces (Goetz et al., 2014). Further, amygdala volume increases have been associated with aggressive behavior among substance users (Schiffer et al., 2011). Collectively these findings suggest that AAS could increase amygdala volume and possibly catalyze or enable aggression behaviors. The hippocampus is involved in spatial memory processes (Squire, 1992). In rats, AAS induce hippocampal apoptosis (Ma and Liu, 2015, Tugyan et al., 2013), and inhibit hippocampal neurogenesis (Brannvall et al., 2005, Novaes Gomes et al., 2014), suggesting that AAS could reduce hippocampal volume, which could be a basis for the AAS-associated spatial memory impairments observed in human and animal studies. The dACC is a cognitive control region involved in attentional processes (Bush and Shin, 2006), which as noted above are abnormal in human AAS users (Hildebrandt et al., 2014). Abnormal dACC activation has been documented in alcohol-dependent subjects performing a spatial working memory fMRI task (Vollstädt-Klein et al., 2010), suggesting that visuospatial dysfunction among AAS users could be related to dACC dysfunction.

Although case reports have documented cerebrovascular problems associated with human AAS use (Akhter et al., 1994, Shimada et al., 2012), no systematic neuroimaging studies have yet assessed human brain effects of long-term AAS use. Accordingly, we acquired from long-term AAS users and nonusers 3 T structural magnetic resonance imaging (MRI), resting state functional connectivity (rsFC) MRI (which maps brain regions thought to be functionally coupled and inherently organized at rest (Greicius, 2008)), and proton magnetic resonance spectroscopy (MRS, which evaluates neurochemistry). We also administered the two computerized tests of visuospatial memory revealing deficits in AAS users in our prior study (Kanayama et al., 2013). Because it is technically challenging to acquire high-quality 3 T MRS spectra from the small, irregularly-shaped amygdala and hippocampus, due in part to partial volume effects (contamination by adjacent structures), we acquired MRS spectra from the dACC, from which MRS data be more reliably acquired. Also, as discussed above, this region may contribute to deficits associated with AAS use. We used MRI to determine whether long-term AAS use is associated with abnormal amygdala and hippocampal volumes and connectivities. Also, given rodent studies suggesting that AAS may enhance the effects of glutamate neurotransmission (Kalinine et al., 2014, Orlando et al., 2007), we used MRS to determine whether long-term AAS use is associated with dACC glutamate abnormalities.

Section snippets

Participants

Study participants were drawn from a pool of about 150 experienced male weightlifters aged 35–55 who had been evaluated in 2011–2014 in a large ongoing study of the cardiac effects of long-term AAS use (Weiner et al., 2013). Since virtually all AAS users are weightlifters, we initially recruited these men by advertising in gymnasiums frequented by AAS users and nonusers (Kanayama et al., 2003, Pope et al., 2012). Participants received a comprehensive interview covering athletic, medical, and

Demographic measures and cognitive test findings

The groups were well matched on demographic variables (Table 1). AAS users reported usage patterns similar to those of long-term AAS users evaluated in our prior studies (Kanayama et al., 2003, Pope et al., 2012, Pope and Katz, 1994; see Table 1), involving about 80% injectable AAS (e.g., testosterone, nandrolone, boldenone) and 20% oral preparations (e.g., methandienone, methenolone, stanozolol). Some participants reported current use of prescription psychoactive medications, including 3 AAS

Discussion

In this brain imaging study, long-term AAS users showed (1) markedly increased right amygdala volumes; (2) markedly decreased right amygdala rsFC, and (3) reduced dACC gln/glu and scyllo-inositol levels compared to nonusers. We also found group differences approaching statistical significance on a visuospatial memory test. To our knowledge, these data represent the first report of brain structural and functional effects of long-term human AAS use.

Role of funding source

The sponsors did not have any role in study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the paper for publication.

Contributors

All authors made: (1) substantial contributions to the conception and analysis and interpretation of data; (2) drafting the article and revising it critically for important intellectual content; and (3) gave approval of the version to be published.

Drs. Pope and Kanayama and Mr. Kerrigan oversaw subject recruitment and screening. Drs. Pope, Kanayama, Kaufman, Jensen, and Mr. Kerrigan oversaw data acquisition and management. Drs. Hudson, Pope, and Janes oversaw statistical analyses. Drs. Kaufman,

Conflict of interest

Dr. Pope has testified approximately once per year in legal cases involving anabolic-androgenic steroids. Dr. Hudson has received grant support from Genentech and Shire; and has received consulting fees from Genentech, Roche, and Shire. All other authors declare that they have no conflicts of interest.

Acknowledgements

Funding for this study was provided in part by NIDA grants R01 DA-29141 (to Drs. Hudson, Kanayama, and Pope) and K01 DA-029645 (to Dr. Janes), and by NIMH grant K23 MH-092397 (to Dr. Brennan).

References (89)

  • E. Kalinine et al.

    Nandrolone-induced aggressive behavior is associated with alterations in extracellular glutamate homeostasis in mice

    Horm. Behav.

    (2014)
  • G. Kanayama et al.

    Anabolic steroid abuse among teenage girls: an illusory problem?

    Drug Alcohol Depend.

    (2007)
  • G. Kanayama et al.

    Long-term psychiatric and medical consequences of anabolic-androgenic steroid abuse: a looming public health concern?

    Drug Alcohol Depend.

    (2008)
  • G. Kanayama et al.

    Cognitive deficits in long-term anabolic-androgenic steroid users

    Drug Alcohol Depend.

    (2013)
  • G. Kanayama et al.

    Risk factors for anabolic-androgenic steroid use among weightlifters: a case-control study

    Drug Alcohol Depend.

    (2003)
  • R.E. Kelly et al.

    Visual inspection of independent components: defining a procedure for artifact removal from fMRI data

    J. Neurosci. Method.

    (2010)
  • E.M. Kouri et al.

    Increased aggressive responding in male volunteers following the administration of gradually increasing doses of testosterone cypionate

    Drug Alcohol Depend.

    (1995)
  • A.Y. Lai et al.

    Scyllo-inositol promotes robust mutant Huntingtin protein degradation

    J. Biol. Chem.

    (2014)
  • C.S. Lynch et al.

    Dihydrotestosterone and estrogen regulation of rat brain androgen-receptor immunoreactivity

    Physiol. Behav.

    (2000)
  • F. Ma et al.

    17beta-trenbolone, an anabolic-androgenic steroid as well as an environmental hormone, contributes to neurodegeneration

    Toxicol. Appl. Pharmacol.

    (2015)
  • K. Magnusson et al.

    Nandrolone decanoate administration elevates hippocampal prodynorphin mRNA expression and impairs Morris water maze performance in male rats

    Neurosci. Lett.

    (2009)
  • J. McLaurin et al.

    Inositol stereoisomers stabilize an oligomeric aggregate of Alzheimer amyloid beta peptide and inhibit abeta -induced toxicity

    J. Biol. Chem.

    (2000)
  • C.S. Menard et al.

    Up-regulation of androgen receptor immunoreactivity in the rat brain by androgenic-anabolic steroids

    Brain Res.

    (1993)
  • F.G. Novaes Gomes et al.

    The beneficial effects of strength exercise on hippocampal cell proliferation and apoptotic signaling is impaired by anabolic androgenic steroids

    Psychoneuroendocrinology

    (2014)
  • R. Orlando et al.

    Nanomolar concentrations of anabolic-androgenic steroids amplify excitotoxic neuronal death in mixed mouse cortical cultures

    Brain Res.

    (2007)
  • H. Pope et al.

    Risk factors for illicit anabolic-androgenic steroid use in male weightlifters: a cross-sectional cohort study

    Biol. Psychiatry

    (2012)
  • A.K. Roy et al.

    Functional connectivity of the human amygdala using resting state fMRI

    NeuroImage

    (2009)
  • F. Tanehkar et al.

    Voluntary exercise does not ameliorate spatial learning and memory deficits induced by chronic administration of nandrolone decanoate in rats

    Horm. Behav.

    (2013)
  • K. Tugyan et al.

    Neuroprotective effect of erythropoietin on nandrolone decanoate-induced brain injury in rats

    Neurosci. Lett.

    (2013)
  • W.R. Yates et al.

    Psychosexual effects of three doses of testosterone cycling in normal men

    Biol. Psychiatry

    (1999)
  • J.W. Younger et al.

    Prescription opioid analgesics rapidly change the human brain

    Pain

    (2011)
  • J. Akhter et al.

    Cerebrovascular accident associated with anabolic steroid use in a young man

    Neurology

    (1994)
  • K.M. Beaver et al.

    Anabolic-androgenic steroid use and involvement in violent behavior in a nationally representative sample of young adult males in the United States

    Am. J. Public Health

    (2008)
  • M. Boldrini et al.

    Hippocampal granule neuron number and dentate gyrus volume in antidepressant-treated and untreated major depression

    Neuropsychopharmacology

    (2013)
  • K. Brannvall et al.

    19-Nortestosterone influences neural stem cell proliferation and neurogenesis in the rat brain

    Eur. J. Neurosci.

    (2005)
  • B.P. Brennan et al.

    Rapid enhancement of glutamatergic neurotransmission in bipolar depression following treatment with riluzole

    Neuropsychopharmacology

    (2010)
  • G. Bush et al.

    The Multi-Source Interference Task: an fMRI task that reliably activates the cingulo-frontal-parietal cognitive/attention network

    Nat. Protoc.

    (2006)
  • Cambridge Cognition

    CANTAB Bibliography

    (2015)
  • F. Caraci et al.

    Neurotoxic properties of the anabolic androgenic steroids nandrolone and methandrostenolone in primary neuronal cultures

    J. Neurosci. Res.

    (2011)
  • P.Y. Choi et al.

    Violence toward women and illicit androgenic-anabolic steroid use

    Ann. Clin. Psychiatry

    (1994)
  • B.M. Cooke et al.

    A brain sexual dimorphism controlled by adult circulating androgens

    Proc. Natl. Acad. Sci. U.S.A.

    (1999)
  • C.D. Fowler et al.

    Newly proliferated cells in the adult male amygdala are affected by gonadal steroid hormones

    J. Neurobiol.

    (2003)
  • M. Greicius

    Resting-state functional connectivity in neuropsychiatric disorders

    Curr. Opin. Neurol.

    (2008)
  • T. Hildebrandt et al.

    The influence of age of onset and acute anabolic steroid exposure on cognitive performance, impulsivity, and aggression in men

    Psychol. Addict. Behav.

    (2014)
  • Cited by (0)

    View full text