Zebrafish models to study drug abuse-related

advertisement
Zebrafish models to study drug abuse-related phenotypes
Adam Stewart*, Keith Wong*, Jonathan Cachat, Siddharth Gaikwad, Evan Kyzar,
Nadine Wu, Peter Hart, Valerie Piet, Eli Utterback, Marco Elegante,
David Tien, Allan V. Kalueff**
Department of Pharmacology and Neuroscience Program, Zebrafish Neuroscience Research
Consortium (ZNRC), Tulane University Medical School, 1430 Tulane Ave., New Orleans, LA
70112, USA
*Both authors contributed equally
**Corresponding Author: Allan V. Kalueff, PhD
Department of Pharmacology, Room SL-83, Tulane University Medical School,
1430 Tulane Ave., New Orleans, LA 70112, USA.
Tel.: +1 504 988 3354. Email: avkalueff@gmail.com
1 Synopsis
Mounting evidence implicates the zebrafish (Danio rerio) as a promising model species for
reward and addiction research. Modeling drug abuse-related behavior in both adult and larval
zebrafish produced a wealth of clinically translatable data, also demonstrating their sensitivity to
various drugs of abuse and the ability to develop tolerance. Several studies have also applied
withdrawal paradigms to model the adverse effects of drug abuse in zebrafish. Here we summarize
recent findings of a wide spectrum of zebrafish drug abuse-related behavioral and physiological
phenotypes, discuss the existing challenges, and outline potential future directions of research in this
field.
Key words: zebrafish, drug abuse, withdrawal, tolerance, anxiety, stress, cortisol
2 Introduction
Drug abuse and addiction are serious mental health and societal problems [1-4]. They
represent complex brain disorders with multiple symptoms (Fig. 1) caused by both genetic and
environmental factors [5-9]. Various experimental (animal) models have been introduced to target
different aspects of drug abuse [10-14].
Zebrafish (Danio rerio) continue to emerge as a new promising model for reward and
addiction research [15-18]. Dopaminergic projections to zebrafish forebrain parallel the mesolimbic
system (implicated in drug addiction in mammals [19]), representing a pathway that is highly
conserved and develops at early ontogenesis [20]. Various behavioral paradigms have been
particularly useful in zebrafish addiction research (Table 1). For example, conditioned place
preference (CPP) and similar models reveal strong rewarding properties of different drugs in
zebrafish [18, 21]. Genetic factors also contribute to zebrafish behavioral responses [22],
demonstrating the link between individual genes and reward phenotypes (e.g., [18, 23]).
Modeling zebrafish behavior traditionally utilizes both adults and younger animals, including
larvae and older, free-feeding fry. While the distinction between larvae and fry is important, for the
purposes of this article, we will apply the term ‘larvae’ to both these stages. Overall, zebrafish larvae
display robust drug-evoked neurobehavioral phenotypes [24], and offer the ability to study multiple
animals simultaneously, within a high-throughput battery [24-26]. However, larval models do not
always display the complex behavior of their adult counterparts, and lack fully established mediatory
and endocrine systems [27], neural circuits [28] and neuromuscular systems [29]. Therefore, both
models should be used complementarily to study drug abuse-related neurobehavioral domains (Fig.
1).
3 A special attention must be paid to selecting the drug concentrations for zebrafish studies.
First, background literature is lacking for many psychotropic drugs, since the zebrafish is a new
model in behavioral pharmacology [30-32]. Second, although zebrafish possess all major
‘mammalian’ neurotransmitters, peptides and hormones [22, 33], the species differences in animal
physiology play a role. For example, unlike humans and rodents, zebrafish have have two forms of
serotonin transporter (SERT A and B) [34-36]. The blood-brain barrier of teleost fish is less
exclusive than in mammals, enabling serotonin to pass through it and affect both central and
peripheral mechanisms [37-39]. Thus, zebrafish may be differentially sensitive to various
serotonergic drugs, compared to other model species. Third, discrepancies are likely within different
zebrafish studies, since it is difficult to translate drug concentrations from larval into adult fish
models. Finally, because the method of drug treatment in most zebrafish studies is immersion
(instead of injection, as in other species), the effective drug concentrations may be based on different
pharmacokinetics than in rodents or humans. However, this does not necessarily represent a flaw of
a model, since water immersion may be advantageous for some drugs (e.g., that are quickly
metabolized or require prolonged treatment) and does not involve injection stress that may confound
behavioral data.
Sensitivity of zebrafish to drugs of abuse
Animal sensitivity to acute and chronic drugs of abuse (including both reward-related and
other behavioral effects) is an important phenotype (Fig. 1) [15-17, 40] which correlates with the
drug’s abuse potential. The sensitivity to drugs of abuse revealed strong genetic determinants of both
increased [45-47] and decreased [42-44] risks of drug abuse. Both animal and clinical models reveal
striking parallels in their sensitivity to cocaine [48], amphetamine [49], benzodiazepines [50],
ethanol [51], nicotine [52], opiates [53], and other psychotropic compounds [54, 55]. Zebrafish
4 models are also sensitive to a wide range of psychotropic compounds (Table 1). While these
responses will be only briefly discussed here, they generally parallel rodent and clinical
observations, thereby confirming the translational value of zebrafish models.
Larval zebrafish display an overt morphine preference, which is reduced by pre-treatment
with naloxone and abolished by blocking dopamine signaling with D1 antagonists [56]. Adult
zebrafish also show a strong dose-dependent preference for morphine [57], which strikingly
resembles rodent responses to this drug [58-60].
Zebrafish models have been extensively used to study the effects of ethanol. In larvae, acute
exposure to ethanol evokes hyperlocomotion at lower doses and a hypolocomotory effect at higher
doses [61]. Strain differences in larval ethanol responses have also been reported [61, 62]. A similar
U-shaped dose-response curve has been observed in adult zebrafish [16], also showing straindependent variations in their responses to ethanol [63], as well as reduced shoaling and increased
aggressiveness [64]. In contrast, chronic ethanol treatment has an anxiolytic effect on zebrafish
behavior [22] (Fig. 2A), also altering the expression of multiple brain genes, some of which are
implicated in the addiction [21] and similarly affected by ethanol in mammals [51, 65].
Nicotine exposure produces strong effects on zebrafish place preference and learning [21,
66]. Although learning and memory are not specifically addressed here, the established effects of
drugs of abuse that affect cognitive functions give further validity to the zebrafish model of drug
abuse [17, 40, 67, 68]. Chronic nicotine exposure in larval zebrafish leads to reduced swimming and
impairs their startle response [69]. In adult zebrafish, acute administration of nicotine has an
anxiolytic-like effect [70, 71] (also see Fig. 2B) similar to its effect in humans and rodents [52].
While zebrafish show a clear preference for cocaine in the CPP paradigm, there are also
several strains with a decreased sensitivity in this model [68, 72]. Overall, zebrafish CPP models
5 display a substantial similarity to rodent cocaine CPP studies [73]. Adult zebrafish treated acutely
with mild doses of cocaine display arousal (e.g., circling, fin extension), increased aggressiveness,
and decreased visual sensitivity [72]. However, higher concentrations of cocaine reduce fish
responses [72] in spite of the high brain cocaine levels [68].
The sensitivity of larval zebrafish to amphetamine [74] generally parallels a similar
locomotor response observed in mammals. While low concentrations of amphetamine increase
activity, higher concentrations of this drug markedly reduce zebrafish locomotion [17, 18]. The
rewarding properties of amphetamine have been reported in adult zebrafish in the CPP test [17], and
also parallel those seen in rodents [49].
Benzodiazepines are known to activate the reward system in rodents [50], and have also been
tested in zebrafish models. For example, both chlordiazepoxide and diazepam display anxiolytic-like
effects in adult zebrafish. While chlordiazepoxide increases exploratory behavior in the light/dark
box paradigm, it does not affect vertical localization in the novel tank test [75, 76]. In contrast,
diazepam increases exploration in the novel tank, exhibiting a biphasic response for low to moderate
doses [75].
Unlike their extensive use in rodent research, hallucinogenic drugs have only recently been
tested in zebrafish. For example, salvinorin A, one of the most potent hallucinogens, exhibits
rewarding properties in the CPP, accelerates zebrafish swimming in acute low doses, and reduces
locomotion (evoking low-velocity "trance-like" state) at high doses [77]. Recently resurrected
interest in psychedelic drug research [55, 78, 79] guided our group’s interest to testing these drugs in
zebrafish. Similar to other fish species’ responses to lysergic acid diethylamide (LSD) [80-83], adult
zebrafish produced significantly shorter latency to enter the top and fewer freezing bouts [67]. LSD
also caused significantly more top transitions and time spent in top, as well as elevated cortisol
6 levels, consistent with its well-known endocrine effects in humans [84] and animals [85, 86]. Acute
administration of another hallucinogen, 3,4-methylenedioxymethamphetamine (MDMA) produced
less clear-cut behaviors (Fig. 3B), albeit showing a similar reduction in freezing (data no shown).
While this may be relevant to known clinical effects of MDMA and LSD (including the
rewarding/euphoric behaviors [54, 55] and elevated corticoids [84-88]), further research is needed to
examine zebrafish sensitivity to various hallucinogenic drugs.
Zebrafish can also be used to study plant compounds with potential psychoactive properties.
For example, kratom (Mitragyna speciosa) is a medicinal leaf, often used as a tea for its calming and
energizing effects. Mitragynine, the major alkaloid identified from kratom, is a partial opioid agonist
producing similar effects to morphine in mammals [89]. When administered acutely, kratom extract
has a mild sedative effect in zebrafish novel tank (Fig. 4), but did not have psychoactive/anxiolytic
action over a wide range of concentrations (1-12 g/L) tested in our laboratory. This suggests that
zebrafish may display differential sensitivity to the drugs’ complex behavioral profiles.
Tolerance and withdrawal
Commonly observed for drugs of abuse in both clinical [90, 91] and animal [40, 92] studies,
tolerance is the progressive reduction of drug sensitivity, which requires higher doses to obtain the
same effects. Tolerance represents an important drug abuse-related phenotype (Fig. 1), mediated by
the brain’s adaptive mechanisms [93-95]. Recent studies have confirmed tolerance in adult zebrafish,
reporting that after chronic exposure to ethanol, zebrafish have a reduced response to the acute
effects of the drug [40]. Another study reported tolerance following chronic ethanol exposure, which
was also influenced by zebrafish genotype (strain) [96]. Tolerance is also seen in zebrafish following
chronic exposure to nicotine [21], collectively paralleling known rodent and clinical findings.
7 Withdrawal is another key phenotype associated with drug abuse [15] (Fig. 1), extensively
studied in various rodents following cessation of ethanol [97], cocaine [98], benzodiazepines [99],
and opiates [100]. These symptoms are also sensitive to various pharmacological [101, 102], genetic
[103] and behavioral [104, 105] factors. Withdrawal is believed to be mediated by the body’s
tendency to maintain homeostasis, in which counter-regulatory mechanisms produce unopposed
effects when a drug is abruptly removed [95, 106-110].
Researchers have recently turned their attention to studying the withdrawal phenomena in
zebrafish (Table 1). Acute discontinuation of drug treatment – acute (single) withdrawal – is a
common form of withdrawal, evoking strong behavioral effects in humans and rodents [111-115]. In
all studies, the most common behavioral manifestations of withdrawal include anxiety, seizures,
sedation, and pain [90, 116-119]. In zebrafish, acute ethanol discontinuation decreases zebrafish
shoaling behavior [120], whereas cocaine withdrawal evokes marked hyperlocomotion marked by
erratic movements and increased exploratory behavior [68, 121]. Acute discontinuation of ethanol,
diazepam, and morphine produces robust anxiogenic-like behavioral responses in zebrafish such as
hypolocomotion, decreased top transitions, and increased freezing bouts in the novel tank [15].
This novelty-based test [22, 71, 122] is an effective paradigm for observing withdrawal
behavior in zebrafish due to these clear-cut anxiety-like responses (see other papers in this issue for
details on zebrafish models of anxiety). Our experiments with chlordiazepoxide (CDP), a sedative
benzodiazepine known to produce anxiolytic effects in both rodents [123] and zebrafish [76], have
tested its withdrawal potential in zebrafish. Two groups of fish (n=15) were administered chronic
CDP (10 mg/L) for 4 months, with another groups of similar size used as a drug-free control. One of
the CDP-treated groups was withdrawn from CDP for 7 days before testing in the novel tank test
(Fig. 5). The withdrawal group displayed an increased latency to the top half of the tank, less
8 transitions to the top, a longer duration in the top, compared to controls. The withdrawal group also
exhibited an increased freezing duration, compared to both the control and the chronic CDP groups
(Fig. 5). Similar to the response observed in both humans [124-126] and rodents [127, 128],
withdrawal elevates zebrafish cortisol, correlating with the expected higher levels of stress and
anxiety [15] (also see Fig. 5 for CDP data).
In humans, chronic drug abuse represents a cyclical process of repeated reward and
withdrawal. Therefore, to more accurately model clinical withdrawal phenomena, repeated
withdrawal models are needed in addition to acute withdrawal studies. Repeated drug withdrawal
paradigms have been recently developed for rodents, showing that both the rat and human share
common triggers of relapse (such as the drug of abuse, stress, stimuli, or the environment
conditioned to the drug of abuse), and that withdrawal selectively potentiates responses to
anxiogenic stimuli [115, 129-132]. A recent study successfully applied repeated withdrawal protocol
to adult zebrafish, reporting that morphine and ethanol withdrawal leads to robust anxiety-like
behaviors [15]. Interestingly, not all drugs of abuse evoke withdrawal. For example, LSD (known for
its low ability to induce withdrawal in humans [133] and animals [134]) does not evoke withdrawal
in both acute and repeated zebrafish models (data not shown).
The complexity of withdrawal phenotypes [135-142], as well as the difficulty in modeling
withdrawal syndrome in animals [143-145], represent another challenge. Potentially interesting
directions of research may focus on neurochemical alterations, neural circuits, and the long-term
consequences [126, 146-148] of drug withdrawal in zebrafish. The genomic profiling of zebrafish
withdrawal also provides further insights, including altered gene expression in the zebrafish brain
following chronic drug treatment and withdrawal [21, 40, 120]. Sex differences in reported for
zebrafish withdrawal-related behaviors [121] parallel sex differences in human [149] and rodent
9 [117, 150-152] withdrawal responses, therefore increasing populational and construct validity of
these models.
Concluding remarks
Because drug abuse is a serious biomedical and societal problem, a better understanding of
the mechanisms behind drug reward and abuse is needed to develop innovate and more efficient
treatments. Considerable efforts have recently been made to develop reliable and high-throughput
assays for zebrafish behavior [17]. Mounting evidence indicates that zebrafish cognition is complex
and often parallel the abilities of mammals [122]. Since cognitive deficits commonly accompany
drug abuse (Fig. 1) [153-156], the ability to quantify both affective and cognitive phenomena in
zebrafish becomes important to study in relation to drug abuse phenotypes. Therefore, novel
behavioral models focusing on reward, cognitive and affective phenotypes may be needed to more
comprehensively model drug abuse in zebrafish.
We have previously emphasized the importance of promoting both larval and adult zebrafish
research [122]. The rich behavioral repertoire of the former complements the sensitivity and highthroughput nature of the latter [157]. Although the genetic aspects of addiction have long been
studied in mammalian organisms, zebrafish are also an ideal model for the use in forward genetics as
they can be rapidly cloned, have a relatively short generation time, and have large progeny sizes that
facilitate large-scale screens [17]. Further analysis of the molecular and biochemical pathways
underlying pre-existing genetic differences in zebrafish, such as strain-dependent variances, will
advance our understanding of how drug abuse affects the brain [62].
The ease of genetic manipulations and potential for high-throughput screening [21, 158, 159]
represent the strengths of zebrafish-based models. However, zebrafish models of drug abuse have
other important strengths, including behavioral complexity, 3-dimensionality, robustness of drug-
10 evoked phenotypes, and the ability to target a wide spectrum of neurobehavioral phenomena (Fig. 1,
Table 1). Therefore, a more comprehensive picture is needed in the field of zebrafish drug abuse
research, with a fair balance between their behavioral and genetic strengths. The growing availability
of new zebrafish strains through the Zebrafish International Repository Center [160], and current
progress of the Sanger Institute’s zebrafish genome project complements mounting support to
zebrafish behavioral research from the National Science Foundation, and recent cross-NIH programs
to develop new zebrafish genetic models and tools for their phenotyping. Together with multiple
lesser initiatives, such as international Zebrafish Neuroscience Research Consortium, this represents
a positive and promising development in the field.
Finally, we acknowledge the importance of applying cross-species and cross-domain
modeling [122] to drug abuse-related phenotypes (Fig. 1). For example, clinical drug abuse is highly
comorbid with anxiety [161], depression [162], bipolar disorder [163], and psychoses [164].
Therefore, animal models that simultaneously target these comorbidities, may lead to valid clinically
relevant experimental paradigms of drug abuse. Furthermore, since genetic variability influences
addiction sensitivity [62], the overlay between behavioral phenotypes and the propensity for, and the
expression of, addictive behavior must be considered. This approach may also improve the ability of
animal models to mimic the entire pathway of drug addiction, rather than a single point along the
continuum [165]. From this point of view, understanding the pathology of drug abuse and addiction
will benefit from implementation of innovative approaches and novel model species, such as
zebrafish.
Acknowledgement: This study is supported by Tulane University Intramural Research program,
Zebrafish Neuroscience Research Consortium (ZNRC) and LA Board of Regents’ P-Fund. The
11 authors thank J. DiLeo, C. Suciu, M. Hook, D. Carlos, K. Rhymes, K. Chang and L. Grossman for
their help with this study.
12 References
1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. Larson, A. and G. Bammer, Why? Who? How? Estimating numbers of illicit drug users: lessons from a case study from the Australian Capital Territory. Aust N Z J Public Health, 1996. 20(5): p. 493‐9. Brady, J.E., et al., Estimating the prevalence of injection drug users in the U.S. and in large U.S. metropolitan areas from 1992 to 2002. J Urban Health, 2008. 85(3): p. 323‐51. Aceijas, C., et al., Estimates of injecting drug users at the national and local level in developing and transitional countries, and gender and age distribution. Sex Transm Infect, 2006. 82 Suppl 3: p. iii10‐
17. Banken, J.A., Drug abuse trends among youth in the United States. Ann N Y Acad Sci, 2004. 1025: p. 465‐71. Hamilton, I., Ensuring integrated treatment for people with mental health and substance use problems. Nurs Times. 106(11): p. 12‐5. Busto, A., et al., Cocaine and amphetamine regulated transcript (CART) gene in the comorbidity of schizophrenia with alcohol use disorders and nicotine dependence. Prog Neuropsychopharmacol Biol Psychiatry. Cheung, J.T., et al., Anxiety and mood disorders and cannabis use. Am J Drug Alcohol Abuse. 36(2): p. 118‐22. Sareen, J., et al., Does a U‐shaped relationship exist between alcohol use and DSM‐III‐R mood and anxiety disorders? J Affect Disord, 2004. 82(1): p. 113‐8. Brunette, M.F., et al., Benzodiazepine use and abuse among patients with severe mental illness and co‐occurring substance use disorders. Psychiatr Serv, 2003. 54(10): p. 1395‐401. Crabbe, J.C., J.K. Belknap, and K.J. Buck, Genetic animal models of alcohol and drug abuse. Science, 1994. 264(5166): p. 1715‐23. Jentsch, J.D., Impulsivity in Animal Models for Drug Abuse Disorders. Drug Discov Today Dis Models, 2008. 5(4): p. 247‐250. Markou, A., et al., Animal models of drug craving. Psychopharmacology (Berl), 1993. 112(2‐3): p. 163‐82. Brady, J.V., Animal models for assessing drugs of abuse. Neurosci Biobehav Rev, 1991. 15(1): p. 35‐
43. Friedman, H. and T.K. Eisenstein, Neurological basis of drug dependence and its effects on the immune system. J Neuroimmunol, 2004. 147(1‐2): p. 106‐8. Cachat, J., et al., Modeling withdrawal syndrome in zebrafish. Behav Brain Res, 2010. 208(2): p. 371‐
6. Gerlai, R., et al., Drinks like a fish: zebra fish (Danio rerio) as a behavior genetic model to study alcohol effects. Pharmacol Biochem Behav, 2000. 67(4): p. 773‐82. Ninkovic, J. and L. Bally‐Cuif, The zebrafish as a model system for assessing the reinforcing properties of drugs of abuse. Methods, 2006. 39(3): p. 262‐74. Webb, K.J., et al., Zebrafish reward mutants reveal novel transcripts mediating the behavioral effects of amphetamine. Genome Biol, 2009. 10(7): p. R81. E. Rink and M.F. Wullimann, Development of the catecholaminergic system in the early zebrafish brain: an immunohistochemical study. Brain Res Dev Brain Res, 2002. 137(1): p. 89‐100. Boehmler, W., et al., Evolution and expression of D2 and D3 dopamine receptor genes in zebrafish. Dev Dyn, 2004. 230(3): p. 481‐93. Kily, L.J., et al., Gene expression changes in a zebrafish model of drug dependency suggest conservation of neuro‐adaptation pathways. J Exp Biol, 2008. 211(Pt 10): p. 1623‐34. Egan, R.J., et al., Understanding behavioral and physiological phenotypes of stress and anxiety in zebrafish. Behav Brain Res, 2009. 205(1): p. 38‐44. 13 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. Ninkovic, J., et al., Genetic identification of AChE as a positive modulator of addiction to the psychostimulant D‐amphetamine in zebrafish. J Neurobiol, 2006. 66(5): p. 463‐75. Best, J.D. and W.K. Alderton, Zebrafish: An in vivo model for the study of neurological diseases. Neuropsychiatr Dis Treat, 2008. 4(3): p. 567‐76. Best, J.D., et al., Non‐associative learning in larval zebrafish. Neuropsychopharmacology, 2008. 33(5): p. 1206‐15. Creton, R., Automated analysis of behavior in zebrafish larvae. Behav Brain Res, 2009. 203(1): p. 127‐36. Kimmel, C.B., et al., Stages of embryonic development of the zebrafish. Dev Dyn, 1995. 203(3): p. 253‐310. Kastenhuber, E., et al., Genetic dissection of dopaminergic and noradrenergic contributions to catecholaminergic tracts in early larval zebrafish. J Comp Neurol. 518(4): p. 439‐58. Dou, Y., M. Andersson‐Lendahl, and A. Arner, Structure and function of skeletal muscle in zebrafish early larvae. J Gen Physiol, 2008. 131(5): p. 445‐53. Rubinstein, A.L., Zebrafish assays for drug toxicity screening. Expert Opin Drug Metab Toxicol, 2006. 2(2): p. 231‐40. Zon, L.I. and R.T. Peterson, In vivo drug discovery in the zebrafish. Nat Rev Drug Discov, 2005. 4(1): p. 35‐44. Liang, A., [Zebrafish‐‐useful model for pharmacodynamics and toxicity screening of traditional Chinese medicine]. Zhongguo Zhong Yao Za Zhi, 2009. 34(22): p. 2839‐42. Mueller, K.P. and S.C. Neuhauss, Behavioral neurobiology: how larval fish orient towards the light. Curr Biol, 2010. 20(4): p. R159‐61. Norton, W.H., A. Folchert, and L. Bally‐Cuif, Comparative analysis of serotonin receptor (HTR1A/HTR1B families) and transporter (slc6a4a/b) gene expression in the zebrafish brain. J Comp Neurol, 2008. 511(4): p. 521‐42. Wang, Y., et al., Characterization and expression of serotonin transporter genes in zebrafish. Tohoku J Exp Med, 2006. 208(3): p. 267‐74. Severinsen, K., et al., Characterisation of the zebrafish serotonin transporter functionally links TM10 to the ligand binding site. J Neurochem, 2008. 105(5): p. 1794‐805. Khan, N. and P. Deschaux, Role of serotonin in fish immunomodulation. J Exp Biol, 1997. 200(Pt 13): p. 1833‐8. Genot, G., R. Morfin, and C. Peyraud, Blood‐brain barrier for serotonin in the eel (Anguilla anguilla L.). Comp Biochem Physiol C, 1981. 68C(2): p. 247‐50. Stoddard, P.K., M.R. Markham, and V.L. Salazar, Serotonin modulates the electric waveform of the gymnotiform electric fish Brachyhypopomus pinnicaudatus. J Exp Biol, 2003. 206(Pt 8): p. 1353‐62. Gerlai, R., V. Lee, and R. Blaser, Effects of acute and chronic ethanol exposure on the behavior of adult zebrafish (Danio rerio). Pharmacol Biochem Behav, 2006. 85(4): p. 752‐61. Balster, R.L., Drug abuse potential evaluation in animals. Br J Addict, 1991. 86(12): p. 1549‐58. Krall, C.M., et al., Marked decrease of LSD‐induced stimulus control in serotonin transporter knockout mice. Pharmacol Biochem Behav, 2008. 88(3): p. 349‐57. Trigo, J.M., et al., 3,4‐methylenedioxymethamphetamine self‐administration is abolished in serotonin transporter knockout mice. Biol Psychiatry, 2007. 62(6): p. 669‐79. Thomsen, M., et al., Dramatically decreased cocaine self‐administration in dopamine but not serotonin transporter knock‐out mice. J Neurosci, 2009. 29(4): p. 1087‐92. Martin, M., et al., Cocaine, but not morphine, induces conditioned place preference and sensitization to locomotor responses in CB1 knockout mice. Eur J Neurosci, 2000. 12(11): p. 4038‐46. Michna, L., et al., Altered sensitivity of CD81‐deficient mice to neurobehavioral effects of cocaine. Brain Res Mol Brain Res, 2001. 90(1): p. 68‐74. 14 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. Lindemann, L., et al., Trace amine‐associated receptor 1 modulates dopaminergic activity. J Pharmacol Exp Ther, 2008. 324(3): p. 948‐56. Reichel, C.M. and R.A. Bevins, Competition between novelty and cocaine conditioned reward is sensitive to drug dose and retention interval. Behav Neurosci, 2010. 124(1): p. 141‐51. Mathews, I.Z., M.D. Morrissey, and C.M. McCormick, Individual differences in activity predict locomotor activity and conditioned place preference to amphetamine in both adolescent and adult rats. Pharmacol Biochem Behav, 2010. 95(1): p. 63‐71. Straub, C.J., W.A. Carlezon, Jr., and U. Rudolph, Diazepam and cocaine potentiate brain stimulation reward in C57BL/6J mice. Behav Brain Res, 2010. 206(1): p. 17‐20. Heilig, M., et al., Acute withdrawal, protracted abstinence and negative affect in alcoholism: are they linked? Addict Biol, 2010. 15(2): p. 169‐84. Jackson, K.J., et al., Characterization of pharmacological and behavioral differences to nicotine in C57Bl/6 and DBA/2 mice. Neuropharmacology, 2009. 57(4): p. 347‐55. Solecki, W., et al., Motivational effects of opiates in conditioned place preference and aversion paradigm‐‐a study in three inbred strains of mice. Psychopharmacology (Berl), 2009. 207(2): p. 245‐
55. Melichar, J.K., M.R. Daglish, and D.J. Nutt, Addiction and withdrawal‐‐current views. Curr Opin Pharmacol, 2001. 1(1): p. 84‐90. Passie, T., et al., The pharmacology of lysergic acid diethylamide: a review. CNS Neurosci Ther, 2008. 14(4): p. 295‐314. Bretaud, S., et al., A choice behavior for morphine reveals experience‐dependent drug preference and underlying neural substrates in developing larval zebrafish. Neuroscience, 2007. 146(3): p. 1109‐16. Lau, B., et al., Dissociation of food and opiate preference by a genetic mutation in zebrafish. Genes Brain Behav, 2006. 5(7): p. 497‐505. Garcia‐Lecumberri, C., et al., Strain differences in the dose‐response relationship for morphine self‐
administration and impulsive choice between Lewis and Fischer 344 rats. J Psychopharmacol, 2010. Sala, M., et al., Dose‐dependent conditioned place preference produced by etonitazene and morphine. Eur J Pharmacol, 1992. 217(1): p. 37‐41. Barr, G.A., W. Paredes, and W.H. Bridger, Place conditioning with morphine and phencyclidine: dose dependent effects. Life Sci, 1985. 36(4): p. 363‐8. Lockwood, B., et al., Acute effects of alcohol on larval zebrafish: a genetic system for large‐scale screening. Pharmacol Biochem Behav, 2004. 77(3): p. 647‐54. Loucks, E. and M.J. Carvan, 3rd, Strain‐dependent effects of developmental ethanol exposure in zebrafish. Neurotoxicol Teratol, 2004. 26(6): p. 745‐55. Gerlai, R., F. Ahmad, and S. Prajapati, Differences in acute alcohol‐induced behavioral responses among zebrafish populations. Alcohol Clin Exp Res, 2008. 32(10): p. 1763‐73. Echevarria, D.J., et al., Does Acute Alcohol Exposure Modulate Aggressive Behaviors in the Zebrafish (Danio rerio), or is the Bark Worse than the Bite? IJCP, 2010. 23(1): p. 62‐69. Sircar, R. and D. Sircar, Repeated ethanol treatment in adolescent rats alters cortical NMDA receptor. Alcohol, 2006. 39(1): p. 51‐8. Levin, E.D. and E. Chen, Nicotinic involvement in memory function in zebrafish. Neurotoxicol Teratol, 2004. 26(6): p. 731‐5. Grossman, L., et al., Characterization of behavioral and endocrine effects of LSD on zebrafish. Behav Brain Res (in press), 2010. Lopez‐Patino, M.A., et al., Anxiogenic effects of cocaine withdrawal in zebrafish. Physiol Behav, 2008. 93(1‐2): p. 160‐71. Parker, B. and V.P. Connaughton, Effects of nicotine on growth and development in larval zebrafish. Zebrafish, 2007. 4(1): p. 59‐68. 15 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. 90. 91. 92. 93. Levin, E.D., Z. Bencan, and D.T. Cerutti, Anxiolytic effects of nicotine in zebrafish. Physiol Behav, 2007. 90(1): p. 54‐8. Levin, E.D., Z. Bencan, and D.T. Cerutti, Assessing stress in zebrafish: Anxiolytic effects of nicotine Neurotoxicology and Teratology, 2006. 28(6): p. 709‐710. Darland, T. and J.E. Dowling, Behavioral screening for cocaine sensitivity in mutagenized zebrafish. Proc Natl Acad Sci U S A, 2001. 98(20): p. 11691‐6. Dietz, D., H. Wang, and M. Kabbaj, Corticosterone fails to produce conditioned place preference or conditioned place aversion in rats. Behav Brain Res, 2007. 181(2): p. 287‐91. Irons, T.D., et al., Acute neuroactive drug exposures alter locomotor activity in larval zebrafish. Neurotoxicol Teratol, 2010. 32(1): p. 84‐90. Bencan, Z., D. Sledge, and E.D. Levin, Buspirone, chlordiazepoxide and diazepam effects in a zebrafish model of anxiety. Pharmacol Biochem Behav, 2009. 94(1): p. 75‐80. Sackerman, J., et al., Zebrafish Behavior in Novel Environments: Effects of Acute Exposure to Anxiolytic Compounds and Choice of Danio rerio Line. Int J Comp Psychol, 2010. 23(1): p. 43‐61. Braida, D., et al., Hallucinatory and rewarding effect of salvinorin A in zebrafish: kappa‐opioid and CB1‐cannabinoid receptor involvement. Psychopharmacology (Berl), 2007. 190(4): p. 441‐8. Gonzalez‐Maeso, J. and S.C. Sealfon, Psychedelics and schizophrenia. Trends Neurosci, 2009. 32(4): p. 225‐32. Dyck, E., Flashback: psychiatric experimentation with LSD in historical perspective. Can J Psychiatry, 2005. 50(7): p. 381‐8. Trout, D.L., Interaction of serotonin and lysergic acid diethylamide in the Siamese fighting fish. J Pharmacol Exp Ther, 1957. 121(1): p. 130‐5. Chessick, R.D., et al., Effect of Pretreatment with Tryptamine, Tryptophan and Dopa on Lsd Reaction in Tropical Fish. Psychopharmacologia, 1964. 5: p. 390‐2. Arbit, J., Effects of LSD‐25 upon Betta splendens: reliability of a bioassay technique. J Appl Physiol, 1957. 10(2): p. 317‐8. Keller, D.L. and W.W. Umbreit, Chemically altered permanent behavior patterns in fish and their cure by reserpine. Science, 1956. 124(3218): p. 407. Bliss, E.L., et al., Reaction of the adrenal cortex to emotional stress. Psychosom Med, 1956. 18(1): p. 56‐76. Sackler, A.M., A.S. Weltman, and H. Owens, Effects of lysergic acid diethylamide on urinary 17‐
ketosteroid and 17‐OH corticosteroid levels of female rats. Nature, 1963. 198: p. 1119‐20. Weltman, A.S. and A.M. Sackler, Metabolic and endocrine effects of lysergic acid diethylamide (LSD‐
25) on male rats. J Endocrinol, 1966. 34(1): p. 81‐90. Parrott, A.C., Cortisol and 3,4‐methylenedioxymethamphetamine: neurohormonal aspects of bioenergetic stress in ecstasy users. Neuropsychobiology, 2009. 60(3‐4): p. 148‐58. de la Torre, R., et al., Pharmacology of MDMA in humans. Ann N Y Acad Sci, 2000. 914: p. 225‐37. Babu, K.M., C.R. McCurdy, and E.W. Boyer, Opioid receptors and legal highs: Salvia divinorum and Kratom. Clin Toxicol (Phila), 2008. 46(2): p. 146‐52. Joseph, E.K., D.B. Reichling, and J.D. Levine, Shared mechanisms for opioid tolerance and a transition to chronic pain. J Neurosci. 30(13): p. 4660‐6. Roberts, J.R. and D. Dollard, Alcohol Levels Do Not Accurately Predict Physical or Mental Impairment in Ethanol‐Tolerant Subjects: Relevance to Emergency Medicine and Dram Shop Laws. J Med Toxicol. Aley, K.O. and J.D. Levine, Different mechanisms mediate development and expression of tolerance and dependence for peripheral mu‐opioid antinociception in rat. J Neurosci, 1997. 17(20): p. 8018‐
23. Popik, P., et al., Human opiorphin: the lack of physiological dependence, tolerance to antinociceptive effects and abuse liability in laboratory mice. Behav Brain Res. 16 94. 95. 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. 113. 114. 115. 116. Wang, Y., et al., Drug‐induced epigenetic changes produce drug tolerance. PLoS Biol, 2007. 5(10): p. e265. Nagy, J., Alcohol Related Changes in Regulation of NMDA Receptor Functions. Curr Neuropharmacol, 2008. 6(1): p. 39‐54. Dlugos, C.A. and R.A. Rabin, Ethanol effects on three strains of zebrafish: model system for genetic investigations. Pharmacol Biochem Behav, 2003. 74(2): p. 471‐80. Morris, S.A., et al., Similar withdrawal severity in adolescents and adults in a rat model of alcohol dependence. Alcohol, 2010. 44(1): p. 89‐98. Santucci, A.C. and A. Rosario, Anxiety‐like responses in adolescent rats following a 10‐11‐day withdrawal period from repeated cocaine administration. Brain Res Bull, 2010. 81(4‐5): p. 441‐4. De Ross, J., et al., Analysis of the chronic intake of and withdrawal from diazepam on emotional reactivity and sensory information processing in rats. Prog Neuropsychopharmacol Biol Psychiatry, 2008. 32(3): p. 794‐802. Becker, G.L., et al., Precipitated and conditioned withdrawal in morphine‐treated rats. Psychopharmacology (Berl), 2010. 209(1): p. 85‐94. Rawls, S.M., D.A. Baron, and J. Kim, beta‐Lactam antibiotic inhibits development of morphine physical dependence in rats. Behav Pharmacol, 2010. 21(2): p. 161‐4. Bhutada, P.S., et al., Inhibitory influence of mecamylamine on ethanol withdrawal‐induced symptoms in C57BL/6J mice. Behav Pharmacol, 2010. 21(2): p. 90‐5. Morice, E., et al., Evidence of long‐term expression of behavioral sensitization to both cocaine and ethanol in dopamine transporter knockout mice. Psychopharmacology (Berl), 2010. 208(1): p. 57‐66. Smith, M.A. and D.L. Yancey, Sensitivity to the effects of opioids in rats with free access to exercise wheels: mu‐opioid tolerance and physical dependence. Psychopharmacology (Berl), 2003. 168(4): p. 426‐34. Saadipour, K., et al., Forced exercise improves passive avoidance memory in morphine‐exposed rats. Pak J Biol Sci, 2009. 12(17): p. 1206‐11. Khong, E., M.G. Sim, and G. Hulse, Benzodiazepine dependence. Aust Fam Physician, 2004. 33(11): p. 923‐6. Tyrer, P.J. and N. Seivewright, Identification and management of benzodiazepine dependence. Postgrad Med J, 1984. 60 Suppl 2: p. 41‐6. Ista, E., et al., Weaning of opioids and benzodiazepines at home after critical illness in infants: a cost‐
effective approach. J Opioid Manag. 6(1): p. 55‐62. Cruz, H.G., et al., Absence and rescue of morphine withdrawal in GIRK/Kir3 knock‐out mice. J Neurosci, 2008. 28(15): p. 4069‐77. Bayard, M., et al., Alcohol withdrawal syndrome. Am Fam Physician, 2004. 69(6): p. 1443‐50. Wiese, J.G., M.G. Shlipak, and W.S. Browner, The alcohol hangover. Ann Intern Med, 2000. 132(11): p. 897‐902. Ashton, H., Benzodiazepine withdrawal: an unfinished story. Br Med J (Clin Res Ed), 1984. 288(6424): p. 1135‐40. Koob, G.F., et al., Opponent process theory of motivation: neurobiological evidence from studies of opiate dependence. Neurosci Biobehav Rev, 1989. 13(2‐3): p. 135‐40. Kokkinidis, L., R.M. Zacharko, and H. Anisman, Amphetamine withdrawal: a behavioral evaluation. Life Sci, 1986. 38(17): p. 1617‐23. Jonkman, S., et al., Spontaneous nicotine withdrawal potentiates the effects of stress in rats. Neuropsychopharmacology, 2008. 33(9): p. 2131‐8. Harris, A.C. and J.C. Gewirtz, Elevated startle during withdrawal from acute morphine: a model of opiate withdrawal and anxiety. Psychopharmacology (Berl), 2004. 171(2): p. 140‐7. 17 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. 127. 128. 129. 130. 131. 132. 133. 134. 135. Strong, M.N., et al., Sex differences in acute ethanol withdrawal severity after adrenalectomy and gonadectomy in Withdrawal Seizure‐Prone and Withdrawal Seizure‐Resistant mice. Alcohol., 2009. 43(5): p. 367‐77. Minozzi, S., et al., Anticonvulsants for alcohol withdrawal. Cochrane Database Syst Rev. 3: p. CD005064. Gowing, L., R. Ali, and J.M. White, Opioid antagonists with minimal sedation for opioid withdrawal. Cochrane Database Syst Rev, 2009(4): p. CD002021. Gerlai, R., et al., Acute and chronic alcohol dose: population differences in behavior and neurochemistry of zebrafish. Genes Brain Behav, 2009. 8(6): p. 586‐99. Lopez Patino, M.A., et al., Gender differences in zebrafish responses to cocaine withdrawal. Physiol Behav, 2008. 95(1‐2): p. 36‐47. Stewart, A., et al., The Developing Utility of Zebrafish in Modeling Neurobehavioral Disorders. Int J Comp Psychol, 2010. 23(1): p. 104‐121. Mathiasen, L.S., N.R. Mirza, and R.J. Rodgers, Strain‐ and model‐dependent effects of chlordiazepoxide, L‐838,417 and zolpidem on anxiety‐like behaviours in laboratory mice. Pharmacol Biochem Behav, 2008. 90(1): p. 19‐36. Bearn, J., et al., Salivary cortisol during opiate dependence and withdrawal. Addict Biol, 2001. 6(2): p. 157‐162. Fox, H.C., E.D. Jackson, and R. Sinha, Elevated cortisol and learning and memory deficits in cocaine dependent individuals: relationship to relapse outcomes. Psychoneuroendocrinology, 2009. 34(8): p. 1198‐207. Shi, J., et al., Time‐Dependent Neuroendocrine Alterations and Drug Craving during the First Month of Abstinence in Heroin Addicts. Am J Drug Alcohol Abuse, 2009: p. 1. Rabbani, M., V. Hajhashemi, and A. Mesripour, Increase in brain corticosterone concentration and recognition memory impairment following morphine withdrawal in mice. Stress, 2009. 12(5): p. 451‐
6. Borlikova, G.G., J. Le Merrer, and D.N. Stephens, Previous experience of ethanol withdrawal increases withdrawal‐induced c‐fos expression in limbic areas, but not withdrawal‐induced anxiety and prevents withdrawal‐induced elevations in plasma corticosterone. Psychopharmacology (Berl), 2006. 185(2): p. 188‐200. Miczek, K.A. and J.A. Vivian, Automatic quantification of withdrawal from 5‐day diazepam in rats: ultrasonic distress vocalizations and hyperreflexia to acoustic startle stimuli. Psychopharmacology (Berl), 1993. 110(3): p. 379‐82. Vorel, S.R., et al., Relapse to cocaine‐seeking after hippocampal theta burst stimulation. Science, 2001. 292(5519): p. 1175‐8. Harris, G.C. and G. Aston‐Jones, Enhanced morphine preference following prolonged abstinence: association with increased Fos expression in the extended amygdala. Neuropsychopharmacology, 2003. 28(2): p. 292‐9. Fendt, M. and R.F. Mucha, Anxiogenic‐like effects of opiate withdrawal seen in the fear‐potentiated startle test, an interdisciplinary probe for drug‐related motivational states. Psychopharmacology (Berl), 2001. 155(3): p. 242‐50. Parsons, J.T., C. Grov, and B.C. Kelly, Club drug use and dependence among young adults recruited through time‐space sampling. Public Health Rep, 2009. 124(2): p. 246‐54. Banerjee, U., Acquisition of conditioned avoidance response in rats under the influence of addicting drugs. Psychopharmacologia, 1971. 22(2): p. 133‐43. Martinotti, G., et al., Alcohol protracted withdrawal syndrome: the role of anhedonia. Subst Use Misuse, 2008. 43(3‐4): p. 271‐84. 18 136. 137. 138. 139. 140. 141. 142. 143. 144. 145. 146. 147. 148. 149. 150. 151. 152. 153. 154. 155. 156. 157. 158. 159. Cooper, Z.D. and M. Haney, Actions of delta‐9‐tetrahydrocannabinol in cannabis: relation to use, abuse, dependence. Int Rev Psychiatry, 2009. 21(2): p. 104‐12. Cruickshank, C.C. and K.R. Dyer, A review of the clinical pharmacology of methamphetamine. Addiction, 2009. Henningfield, J.E., et al., Tobacco dependence and withdrawal: Science base, challenges and opportunities for pharmacotherapy. Pharmacol Ther, 2009. Prat, G., A. Adan, and M. Sanchez‐Turet, Alcohol hangover: a critical review of explanatory factors. Hum Psychopharmacol, 2009. Shoptaw, S.J., et al., Treatment for amphetamine withdrawal. Cochrane Database Syst Rev, 2009(2): p. CD003021. Teixeira, M.Z., Antidepressants, suicidality and rebound effect: evidence of similitude? Homeopathy, 2009. 98(2): p. 114‐21. Wu, L.T., et al., The construct and measurement equivalence of cocaine and opioid dependences: A National Drug Abuse Treatment Clinical Trials Network (CTN) study. Drug Alcohol Depend, 2009. Becker, H.C., Animal models of alcohol withdrawal. Alcohol Res Health, 2000. 24(2): p. 105‐13. Braw, Y., et al., Withdrawal emotional‐regulation in infant rats from genetic animal models of depression. Behav Brain Res, 2008. 193(1): p. 94‐100. Keane, B. and B.E. Leonard, Rodent models of alcoholism: a review. Alcohol Alcohol, 1989. 24(4): p. 299‐309. Nava, F., et al., Relationship between plasma cortisol levels, withdrawal symptoms and craving in abstinent and treated heroin addicts. J Addict Dis, 2006. 25(2): p. 9‐16. Li, S.X., et al., Serum cortisol secretion during heroin abstinence is elevated only nocturnally. Am J Drug Alcohol Abuse, 2008. 34(3): p. 321‐8. Zhang, G.F., et al., Dysfunction of the hypothalamic‐pituitary‐adrenal axis in opioid dependent subjects: effects of acute and protracted abstinence. Am J Drug Alcohol Abuse, 2008. 34(6): p. 760‐8. Fox, H.C., et al., Gender differences in cardiovascular and corticoadrenal response to stress and drug cues in cocaine dependent individuals. Psychopharmacology (Berl), 2006. 185(3): p. 348‐57. Alves, C.J., et al., Hormonal, neurochemical, and behavioral response to a forced swim test in adolescent rats throughout cocaine withdrawal. Ann N Y Acad Sci, 2008. 1139: p. 366‐73. Butler, T.R., et al., Sex Differences in Caffeine Neurotoxicity Following Chronic Ethanol Exposure and Withdrawal. Alcohol Alcohol, 2009. 44(6): p. 567‐74. Taylor, A.N., et al., Sex differences in ethanol‐induced hypothermia in ethanol‐naive and ethanol‐
dependent/withdrawn rats. Alcohol Clin Exp Res, 2009. 33(1): p. 60‐9. Barker, M.J., et al., Persistence of cognitive effects after withdrawal from long‐term benzodiazepine use: a meta‐analysis. Arch Clin Neuropsychol, 2004. 19(3): p. 437‐54. Rogers, P.J., et al., Effects of caffeine and caffeine withdrawal on mood and cognitive performance degraded by sleep restriction. Psychopharmacology (Berl), 2005. 179(4): p. 742‐52. Rapeli, P., et al., Cognitive function during early abstinence from opioid dependence: a comparison to age, gender, and verbal intelligence matched controls. BMC Psychiatry, 2006. 6: p. 9. Kelley, B.J., et al., Cognitive impairment in acute cocaine withdrawal. Cogn Behav Neurol, 2005. 18(2): p. 108‐12. Rihel, J., et al., Zebrafish behavioral profiling links drugs to biological targets and rest/wake regulation. Science, 2010. 327(5963): p. 348‐51. He, S., et al., Genetic and transcriptome characterization of model zebrafish cell lines. Zebrafish, 2006. 3(4): p. 441‐53. Hogan, B.M., et al., Manipulation of gene expression during zebrafish embryonic development using transient approaches. Methods Mol Biol, 2008. 469: p. 273‐300. 19 160. 161. 162. 163. 164. 165. 166. 167. Sprague, J., et al., The Zebrafish Information Network: the zebrafish model organism database provides expanded support for genotypes and phenotypes. Nucleic Acids Res, 2008. 36(Database issue): p. D768‐72. Schneier, F.R., et al., Social anxiety disorder and alcohol use disorder co‐morbidity in the National Epidemiologic Survey on Alcohol and Related Conditions. Psychol Med. 40(6): p. 977‐88. Perkins, K.A., et al., Differences in negative mood‐induced smoking reinforcement due to distress tolerance, anxiety sensitivity, and depression history. Psychopharmacology (Berl). 210(1): p. 25‐34. Swann, A.C., The strong relationship between bipolar and substance‐use disorder. Ann N Y Acad Sci. 1187: p. 276‐93. Gouzoulis‐Mayfrank, E., [Dual diagnosis psychosis and substance use disorders: theoretical foundations and treatment]. Z Kinder Jugendpsychiatr Psychother, 2008. 36(4): p. 245‐53. Warnick, J.E., J.L. LaPorte, and A.V. Kalueff, Domain interplay in mice and men: New possibilities for the “natural kinds” theory of emotion New Ideas in Psychology, 2010. Kily, L., et al., A conditioned place preference assay for modeling addiction in zebrafish. Proc Brit Pharm Soc, 2006. 4(2). Wong, K., et al., Analyzing habituation responses to novelty in zebrafish (Danio rerio). Behav Brain Res, 2010. 208(2): p. 450‐7. 20 Table 1: Sensitivity of zebrafish models to selected drugs of abuse (reported positive findings:
A - adult zebrafish models; L - larval zebrafish models)
Cocaine
A
A
References
LSD*
Nicotine
A
Benzodiazepines
Ethanol
AL
Caffeine
Morphine
Domains
Amphetamine
Drugs
Reward
properties
[17, 21, 56, 57, 72, 166]
A
[16, 22, 56, 57, 61, 62,
Acute behavioral
effects
AL
AL
A
A
A
A
A
A
A
A
AL
AL
A
AL
A
A
67-69, 72, 74-76, 167]
Chronic
behavioral effects
A
[15, 22, 68]
A
Developing
tolerance
[15, 21, 40, 96]
A
Acute (single)
withdrawal
A
A
A
*
A
*
[15, 22, 68, 120]
Repeated
withdrawal
[15], Fig. 5
*Lysergic acid diethylamide (note that its acute or repeated withdrawal in our studies with adult
zebrafish failed to evoke overt anxiety, which is in line with clinically known weak withdrawalevoking potential of this drug).
21 Figure 1: Multiple inter-related domains of drug abuse (note that all these behavioral domains
are highly relevant to zebrafish models of drug abuse described here)
22 Figure 2. Examples of anxiolytic effects of chronic ethanol and acute nicotine in a 6-min novel
tank test. A - Chronic ethanol (0.3% for 1 week; n = 24 for each group). B - Acute nicotine
administration (10 mg/L for 5 min; n = 15 per each group). **P<0.01, ***P<0.001, U-test.
23 Figure 3. Examples of behavioral effects of hallucinogenic drugs in a 6-min novel tank test. A Acute lysergic acid diethylamide (LSD) administration (250 µg/L for 20 min; n = 10 per each
group). B - Acute 3,4-methylenedioxymethamphetamine (MDMA) administration (5 mg/L for
20 min; n = 10 per each group). ***P<0.001, U-test.
24 Figure 4. Examples of behavioral effects of kratom extract in a 6-min novel tank test. Fish
were acutely exposed to 6 g/L of kratom extract for 20 min (n=10 per each group).
25 Figure 5. Behavioral effects of 100 mg/L chronic chlordiazepoxide (CDP) in a 6-min novel tank
test. CDP was administered for 4 months, after which one group was withdrawn for 7 days (n
= 15 per each group). There was significant group effect for latency to upper half (F(2, 44) =
8.7, P<0.001), transitions to top (F(2, 44) = 5.2, P<0.01), time in top (F(2, 44) = 6.1, P<0.01) and
freezing duration (F(2, 44) = 14.7, P<0.001). *P<0.05, **P<0.01, ***P<0.001, #P=0.05-0.1
(trend) between the groups; post-hoc Tukey test for significant one-way ANOVA data.
26 
Download