Elsevier

Biological Psychiatry

Volume 51, Issue 5, 1 March 2002, Pages 400-406
Biological Psychiatry

Original article
Mismatch negativity predicts psychotic experiences induced by nmda receptor antagonist in healthy volunteers

https://doi.org/10.1016/S0006-3223(01)01242-2Get rights and content

Abstract

Background: Previous studies indicate that mismatch negativity (MMN)—a preattentive auditory event-related potential (ERP)–depends on NMDA receptor (NMDAR) functioning. To explore if the strength of MMN generation reflects the functional condition of the NMDAR system in healthy volunteers, we analyzed correlations between MMN recorded before drug administration and subsequent responses to the NMDAR antagonist ketamine or the 5-HT2a agonist psilocybin.

Methods: In two separate studies, MMN was recorded to both frequency and duration deviants prior to administration of ketamine or psilocybin. Behavioral and subjective effects of ketamine and psilocybin were assessed with the Brief Psychiatric Rating Scale and the OAV Scale—a rating scale developed to measure altered states of consciousness. Correlations between ERP amplitudes (MMN, N1, and P2) and drug-induced effects were calculated in each study group and compared between them.

Results: Smaller MMN to both pitch and duration deviants was significantly correlated to stronger effects during ketamine, but not psilocybin administration. No significant correlations were observed for N1 and P2.

Conclusions: Smaller MMN indicates a NMDAR system that is more vulnerable to disruption by the NMDAR antagonist ketamine. MMN generation appears to index the functional state of NMDAR-mediated neurotransmission even in subjects who do not demonstrate any psychopathology.

Introduction

Mismatch negativity (MMN) is an auditory event-related potential (ERP) indexing preattentive detection of stimulus deviance (Näätänen 1995). It is automatically generated with a latency of 100 to 200 ms after the presentation of a stimulus that deviates in one of its physical dimensions (pitch, intensity, duration, location) from preceding frequently repeated standard stimuli Näätänen 1995, Novak et al 1990, Ritter et al 1995. Mismatch negativity is thus the manifestation of a preattentive process that compares the deviant stimulus to the sensory memory trace of the standard stimulus Näätänen 1990, Novak et al 1990, Ritter et al 1995.

Mismatch negativity-like activities can also be recorded in animals and thus are amenable to investigations of its underlying neurobiological substrate Csepe et al 1987, Javitt et al 1994, Javitt et al 1996. Studies in monkeys and cats show that MMN specifically involves activation of the supragranular layer of the primary and secondary auditory cortices (Karmos et al 1998; Javitt et al 1994, Javitt et al 1996). It has been hypothesized that this activation involves excitation of pyramidal neurons as a net result of two processes: 1) reduction of tonic inhibition (disinhibition) due to stimulation by the standard tone, and 2) excitation of NMDA receptor (NMDAR) in response to the deviant stimulus (Schroeder et al 1997; Karmos et al personal communication; Javitt et al 1996). NMDAR functions are uniquely sensitive to membrane potential (Cotman et al 1995); thus, the magnitude of stimulation-induced current flow through NMDAR depends upon the degree of neuronal disinhibition. This property makes the NMDAR uniquely suited for mediating conditional responses such as MMN. Studies in monkeys have indeed demonstrated that intracortical and systemic application of NMDAR antagonists selectively abolishes MMN-like activities without affecting such sensory ERPs as N1 Javitt et al 1996, Javitt et al 1994. In addition, we recently demonstrated that ketamine, a noncompetitive NMDAR antagonist, selectively impairs MMN generation in humans without reducing sensory ERPs (Umbricht et al 2000). Furthermore, ketamine significantly reduced the magnetic counterpart of MMN, MMNm, in healthy volunteers (Sauer et al 2000). The NMDAR antagonist nitrous oxide (N2O) (Jevtovic-Todorovic et al 1998) has also been reported to induce a significant reduction of MMN (Pang et al 1999). In contrast, the psychotomimetic psilocybin does not significantly affect MMN in healthy volunteers, although it reduces N1 (Umbricht et al 2001).

Psilocybin (O-phosphoryl-4-hydroxy-N,N-dimethyltryptamine)—a naturally occurring indoleamine hallucinogen—is readily metabolized to psilocin after ingestion (Hasler et al 1997). In rats, psilocin binds with high affinity to the 5-HT2A (Ki = 6.0 nM) and with moderate affinity to the 5-HT1A receptor (Ki = 190 nM) (McKenna et al 1990). Although indoleamine hallucinogens bind to various subtypes of serotonin receptors, the hallucinogenic and cognitive effects are primarily believed to be mediated through agonist action at the 5-HT2A receptor Aghajanian and Marek 1999a, Vollenweider et al 1998. Moreover, the affinity of indoleamine hallucinogens at the 5-HT2A receptor is highly correlated with their hallucinogenic potency in humans (Aghajanian et al 1999a).

In summary, the available evidence strongly indicates a central role of the NMDAR in MMN generation. Thus, deficits in MMN generation as observed in such neuropsychiatric disorders as schizophrenia Catts et al 1995, Javitt et al 1995, Umbricht et al 1998 may be a manifestation of deficient NMDAR-dependent neurotransmission Olney and Farber 1995, Javitt and Zukin 1991.

Furthermore, these data suggest that even in non-patient subjects the strength of MMN generation may reflect the functional condition of NMDAR systems. To explore this hypothesis, we examined the correlations between MMN recorded prior to drug administration and psychosis-like experiences and behavioral effects induced by the subsequent administration of ketamine or the psychotomimetic psilocybin. Ketamine, and to a lesser extent psilocybin, can induce signs and symptoms in healthy volunteers that mimic some psychotic symptoms of schizophrenia Krystal et al 1994, Vollenweider et al 1998. Thus, we hypothesized that if the strength of MMN indeed reflects the functional condition of the NMDAR system (i.e., its efficiency or redundancy), then the effects of ketamine should be more disruptive. (i.e., stronger in a subject with a smaller MMN at baseline). In other words, an inverse correlation between baseline MMN and ketamine-induced psychosis-like experiences should exist. On the other hand, such a correlation should not be observed between MMN and the effects of psilocybin.

To test this hypothesis, data from two separate studies on the effects of ketamine and psilocybin in healthy volunteers were analyzed. The main goal of both studies was to assess the effects of ketamine or psilocybin, respectively, on MMN generation. The results of these studies have been (Umbricht et al 2000) or will be reported elsewhere (Umbricht unpublished). Here, we only report on the session with active drug administration (ketamine infusion or psilocybin administration, respectively).

Section snippets

Methods and materials

Data from two separate studies of effects of ketamine and psilocybin in healthy volunteers (ketamine study: N = 20, M/F = 14/6, mean age = 24.6 ± 2.9 y; psilocybin study: N = 18, M/F = 10/8, mean age = 25.1 ± 4.3 y) were analyzed. The details of the methodology—identical in many aspects for both studies—have been published previously (Umbricht et al 2000). Both studies had been approved by the ethics committee of the Psychiatric University Hospital Zurich. Briefly, prior to study entry, all

Results

Both ketamine and psilocybin administration induced robust behavioral effects as evidenced in significant increases of the BPRS total and the BPRS psychosis factor scores and strong subjective effects as rated on the OAV questionnaire (see Table 1). The increase of the BPRS total and psychosis factor score was greater during ketamine than psilocybin administration. The difference of the BPRS psychosis factor score was significant (t = 3.15, df = 35, p < .05). However, the subjectively rated

Discussion

Previous studies have demonstrated that the auditory event-related potential MMN is significantly reduced or abolished by NMDAR antagonists in humans and nonhuman primates, indicating a role of the NMDAR in its generation Umbricht et al 2000, Javitt et al 1996, Sauer et al 2000, Pang and Fowler 1999. We recently reported that ketamine significantly reduced MMN in healthy volunteers (Umbricht et al 2000). In this additional study, we explored the hypothesis that MMN in these healthy

Acknowledgements

Supported by grants from the Swiss National Science Foundation to Dr. Umbricht (SNF 32-050957.97) and to Dr. Vollenweider (SNF 3200-040900.94), and the Heffter Research Institute (USA).

References (39)

  • F.X Vollenweider et al.

    Differential psychopathology and patterns of cerebral glucose utilisation produced by (S)- and (R)-ketamine in healthy volunteers using positron emission tomography (PET)

    European Neuropsychopharmacol

    (1997)
  • F.X Vollenweider et al.

    5-HT modulation of dopamine release in basal ganglia in psilocybin-induced psychosis in man - a pet study with [11C]raclopride

    Neuropsychopharmacology

    (1999)
  • F.X Vollenweider et al.

    Effects of (S)-ketamine on striatal dopaminea [11C]raclopride PET study of a model psychosis in humans

    J Psychiatr Res

    (2000)
  • A Anand et al.

    Attenuation of the neuropsychiatric effects of ketamine with lamotriginesupport for hyperglutamatergic effects of N-methyl-D- aspartate receptor antagonists

    Arch Gen Psychiatry

    (2000)
  • I Bodmer et al.

    Aussergewöhnliche Bewusstseinszustände - ihre gemeinsame Struktur und Messung

  • A Breier et al.

    Effects of NMDA antagonism on striatal dopamine release in healthy subjectsapplication of a novel PET approach

    Synapse

    (1998)
  • S.V Catts et al.

    Brain potential evidence for an auditory sensory memory deficit in schizophrenia

    Am J Psychiatry

    (1995)
  • C.W Cotman et al.

    Excitatory Amino Acid Neurotransmission

  • V Csépe et al.

    Effects of signal probability on sensory evoked potentials in cats

    Inter J Neurosci

    (1987)
  • Cited by (208)

    View all citing articles on Scopus
    View full text